Cover by D.Meissner WHO/GRA

advertisement
Cover by D.Meissner WHO/GRA
These Guidelines were adopted by the WHO Expert Committee on Biological
Standardization at its 59th meeting which took place in Geneva from 13 to
17 October 2008 and will be published in the WHO Technical Report Series.
© World Health Organization 2010
All rights reserved. Publications of the World Health Organization can be obtained from WHO Press, World Health
Organization, 20 Avenue Appia, 1211 Geneva 27, Switzerland (tel.: +41 22 791 3264; fax: +41 22 791 4857; e-mail:
bookorders@who.int). Requests for permission to reproduce or translate WHO publications – whether for sale or for
noncommercial distribution – should be addressed to WHO Press, at the above address (fax: +41 22 791 4806;
e-mail: permissions@who.int).
The designations employed and the presentation of the material in this publication do not imply the expression of any
opinion whatsoever on the part of the World Health Organization concerning the legal status of any country, territory,
city or area or of its authorities, or concerning the delimitation of its frontiers or boundaries. Dotted lines on maps
represent approximate border lines for which there may not yet be full agreement.
The mention of specific companies or of certain manufacturers’ products does not imply that they are endorsed or
recommended by the World Health Organization in preference to others of a similar nature that are not mentioned.
Errors and omissions excepted, the names of proprietary products are distinguished by initial capital letters.
All reasonable precautions have been taken by the World Health Organization to verify the information contained in
this publication. However, the published material is being distributed without warranty of any kind, either expressed or
implied. The responsibility for the interpretation and use of the material lies with the reader. In no event shall the
World Health Organization be liable for damages arising from its use.
Contents
1.
Introduction............................................................................................................................
1
2.
List of abbreviations and definitions used ...........................................................................
2
3.
General considerations ..........................................................................................................
7
3.1
3.2
3.3
3.4
3.5
Historical background....................................................................................................
The use of serum versus plasma as source material ......................................................
Antivenom purification methods and product safety.....................................................
Pharmacokinetics and pharmacodynamics of antivenoms.............................................
Need for national and regional reference venom preparations .....................................
7
7
7
8
8
Epidemiological background ................................................................................................
8
4,1
4.2
Global burden of snakebites ..........................................................................................
Main recommendations..................................................................................................
8
9
Worldwide distribution of venomous snakes.......................................................................
10
5.1
5.2
5.3
5.4
5.5
Taxonomy of venomous snakes.....................................................................................
Medically important venomous snakes..........................................................................
Minor venomous snake species .....................................................................................
Sea snake venoms ..........................................................................................................
Main recommendations..................................................................................................
10
13
15
16
16
Antivenoms design: selection of snake venoms ...................................................................
17
6.1
6.2
6.3
Selection and preparation of representative venom mixtures ........................................
Manufacture of monospecific or polyspecific antivenoms ............................................
Main recommendations..................................................................................................
17
17
19
Preparation and storage of snake venom.............................................................................
19
7.1
7.2
7.3
Production of snake venoms for immunization .............................................................
Staff responsible for handling snakes ............................................................................
Main recommendations..................................................................................................
20
25
26
Quality control of venoms .....................................................................................................
27
8.1
8.2
8.3
8.4
Records and traceability.................................................................................................
National reference materials .........................................................................................
Characterization of venom batches................................................................................
Main recommendations..................................................................................................
27
28
28
28
9.
Overview of the production process of antivenoms ............................................................
29
10.
Selection and veterinary health care of animals used for production of antivenoms ......
31
10.1
10.2
10.3
10.4
31
31
31
32
4.
5.
6.
7.
8.
Quarantine period ..........................................................................................................
Veterinary surveillance and vaccinations ......................................................................
Animal health surveillance after inclusion in the herd ..................................................
Main recommendations..................................................................................................
iii
11.
12.
13.
14.
15.
16.
iv
Immunization regimens and use of adjuvant.......................................................................
33
11.1
11.2
11.3
11.4
11.5
11.6
11.7
11.8
11.9
Animals used in antivenom production .........................................................................
Venoms used for immunization .....................................................................................
Preparation of venom doses ...........................................................................................
Detoxification of venom ................................................................................................
Immunological adjuvants...............................................................................................
Preparation of immunogen in adjuvants ........................................................................
Immunization of animals ...............................................................................................
Traceability of the immunization process ......................................................................
Main recommendations..................................................................................................
33
34
34
35
35
35
36
38
39
Collection and control of animal plasma for fractionation ................................................
39
12.1
12.2
12.3
12.4
12.5
12.6
12.7
Health control of the animal prior to and during bleeding sessions...............................
Premises for blood or plasma collection ........................................................................
Blood or plasma collection session................................................................................
Labelling and identification ...........................................................................................
Pooling ...........................................................................................................................
Control of plasma prior to fractionation .......................................................................
Main recommendations..................................................................................................
40
40
40
40
42
43
43
Purification of immunoglobulins and immunoglobulin fragments in the production
of antivenoms..........................................................................................................................
44
13.1
13.2
13.3
13.4
Good manufacturing practices .......................................................................................
Purification of the active substance ...............................................................................
Pharmacokinetic and pharmacodynamic properties of IgG, F(ab')2 and Fab.................
Main recommendations..................................................................................................
44
44
55
56
Control of infectious risks......................................................................................................
56
14.1
14.2
14.3
14.4
14.5
14.6
14.7
Background ....................................................................................................................
Risk of viral contamination of the starting plasma ........................................................
Viral validation of manufacturing processes..................................................................
Viral validation studies of antivenom immunoglobulins ...............................................
Production-scale implementation of process steps contributing to viral safety .............
Transmissible spongiform encephalopathy....................................................................
Main recommendations..................................................................................................
56
57
57
61
65
66
67
Quality control of antivenoms...............................................................................................
68
15.1 Tests ...............................................................................................................................
15.2 Antivenom reference preparations .................................................................................
15.3 Main recommendations..................................................................................................
68
73
73
Stability, storage and distribution of antivenoms................................................................
73
16.1
16.2
16.3
16.4
73
74
74
74
Stability..........................................................................................................................
Storage ...........................................................................................................................
Distribution ....................................................................................................................
Main recommendations..................................................................................................
Preclinical assessment of antivenoms ...................................................................................
75
17.1
17.2
17.3
17.4
17.5
17.6
Introduction ...................................................................................................................
Essential assay for preclinical testing of antivenoms: prevention of lethality ...............
Additional recommended assays for preclinical testing of antivenoms.........................
Development of alternative assays to replace murine lethality testing..........................
Limitations of preclinical assays....................................................................................
Main recommendations..................................................................................................
75
76
76
79
79
80
Clinical assessment of antivenoms........................................................................................
80
18.1
18.2
18.3
18.4
Introduction....................................................................................................................
Clinical studies of antivenoms.......................................................................................
Post-marketing surveillance...........................................................................................
Main recommendations..................................................................................................
80
81
83
85
Role of national regulatory authorities ................................................................................
85
19.1 Impact of good manufacturing practices........................................................................
19.2 Establishment licence and Inspections...........................................................................
86
86
Authors and Acknowledgements ....................................................................................................
88
References .........................................................................................................................................
92
Appendix 1
Worldwide distribution of medically important venomous snakes .............................................
102
Appendix 2
Summary protocol of manufacturing and control of snake antivenom immunoglobulins........
132
17.
18.
19.
v
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
1
INTRODUCTION
The unavailability of effective snake antivenom immunoglobulins (antivenoms) to treat the
specific types of snakebite envenomings encountered in various regions of the world has become
a critical health issue at global level. The crisis has reached its greatest intensity in sub-Saharan
Africa, but other regions, such as south-east Asia, are also suffering from a lack of effective and
affordable products.
The complexity of the production of antivenoms, in particular the importance of preparing
appropriate snake venom mixtures for the production of hyperimmune plasma (the source of
antivenom immunoglobulins), the decreasing number of producers and the fragility of the
production systems in developing countries further jeopardize the availability of effective
antivenoms in Africa, Asia, the Middle East and South America. Most of the remaining current
producers are located in countries where the application of quality and safety standards needs to
be improved.
In October 2005, the WHO Expert Committee on Biological Standardization (ECBS) recognized
the extent of the problem and asked the WHO Secretariat to support and strengthen world
capacity to ensure long-term and sufficient supply of safe and efficient antivenoms. In March
2007, snake antivenom immunoglobulins were included in the WHO Model List of Essential
Medicines (1), acknowledging their role in a primary health care system.
Urgent measures are needed to support the design of immunizing snake venom mixtures that can
be used to make the right polyspecific antivenoms for various geographical areas of the world.
Sustainable availability of effective and safe antivenom immunoglobulins should be ensured and
production systems for these effective treatments should be strengthened at global level.
Meaningful preclinical assessment of the neutralizing capacity of snake antivenom
immunoglobulins needs to be done before these products are used in humans and medicines
regulatory authorities should enforce the licensing of these products before they are used in the
population.
The present “WHO Guidelines for the production, control and regulation of snake antivenoms
immunoglobulins” were developed in response to the above-mentioned needs. These Guidelines
cover all the steps involved in the production, control and regulation of venoms and antivenoms,
as well as an Appendix providing detailed information about the distributions of the most
important snake venoms for use in antivenoms preparation in each country, territory or
geographical area. It is hoped that this document, by covering comprehensively the current
existing experience in the manufacture, preclinical and clinical assessment of these products will
serve as a guide to national control authorities and manufacturers in the support of worldwide
production of these essential medicines. The production of snake antivenoms following good
manufacturing practices should be the aim of all countries involved in the manufacture of these
life-saving biological products.
In addition to the need to produce appropriate antivenoms, other issues that need to be addressed
include ensuring that antivenoms are appropriately used and that outcomes for envenomed
patients are improved. This will entail availability of antivenoms and appropriate distribution
policies, affordability of envenoming treatment and training of health workers to allow safe and
effective use of antivenoms and effective management of snakebite envenomings. These
important issues are beyond the scope of this document and will not be further addressed
specifically here, but should be considered as vital components in the care pathway for
envenoming.
1
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
2
LIST OF ABBREVIATIONS AND DEFINITIONS USED
The definitions given below apply to the terms used in these Guidelines. They may have
different meanings in other contexts.
Antivenoms (also called antivenins)
A purified fraction of immunoglobulins or immunoglobulin fragments fractionated from the
plasma of animals that have been immunized against a snake venom or a snake venom mixture.
Apheresis
Procedure whereby blood is removed from the donor, separated by physical means into
components and one or more of them returned to the donor.
Batch
A defined quantity of starting material or product manufactured in a single process or series of
processes so that it is expected to be homogeneous.
Batch records
All documents associated with the manufacture of a batch of bulk product or finished product.
They provide a history of each batch of product and of all circumstances pertinent to the quality
of the final product.
Blood collection
A procedure whereby a single donation of blood is collected in an anticoagulant and/or
stabilizing solution, under conditions designed to minimize microbiological contamination of the
resulting donation.
Bulk product
Any product that has completed all processing stages up to, but not including, aseptic filling and
final packaging.
Clean area
An area with defined environmental control of particulate and microbial contamination,
constructed and used in such a way as to reduce the introduction, generation, and retention of
contaminants within the area.
Combined antivenoms
Antivenoms directed against several venoms, prepared by mixing different monospecific plasma
prior to the plasma fractionation process, or purified monospecific antivenom fractions prior to
the aseptic filling stage.
Contamination
The undesired introduction of impurities of a microbiological or chemical nature, or of foreign
matter, into or on to a starting material or intermediate during production, sampling, packaging,
or repackaging, storage or transport.
2
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Convention on International Trade in Endangered Species of Wild Fauna and Flora (CITES)
An international agreement between governments. Its aim is to ensure that international trade in
specimens of wild animals and plants does not threaten their survival.
Cross-contamination
Contamination of a starting material, intermediate product or finished product with another
starting material or product during production.
Cross-neutralization
The ability of an antivenom raised against a venom, or a group of venoms, to react and neutralize
the toxic effects of the venom of a related species not included in the immunizing mixture.
Desiccation
A storage process where venoms are dehydrated under vacuum in the presence of calcium salts
or phosphoric acid.
EIA
Enzyme immunoassay.
Envenoming
Process by which venom is injected into a human by the bite of a poisonous snake, leading to
pathological manifestations (also called envenomation).
Fab
A monovalent immunoglobulin fragment resulting from the proteolytic digestion of
immunoglobulins by papain.
F(ab')2
A bivalent immunoglobulin
immunoglobulins by pepsin.
fragment
resulting
from
the
proteolytic
digestion
of
Fractionation
Large-scale process by which animal plasma is separated to isolate the immunoglobulin fraction,
that is further processed for therapeutic use or may be subjected to digestion with pepsin or
papain to generate immunoglobulin fragments. The term fractionation is generally used to
describe a sequence of processes, generally including plasma protein precipitation and/or
chromatography, ultrafiltration and filtration steps.
Freund complete adjuvant (FCA)
An adjuvant that may be used in the immunization process of animals to enhance the immune
response to venoms. It is composed of mineral oil, an emulsifier and inactivated Mycobacterium
tuberculosis.
Freund incomplete adjuvant (FIA)
An adjuvant that may be used in the immunization process of animals to enhance the immune
response to venoms. It is composed of mineral oil and an emulsifier.
3
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Good manufacturing practices (GMP)
That part of quality assurance which ensures that products are consistently produced and
controlled to the quality standards appropriate to their intended use and as required by the
marketing authorization or product specification. It is concerned with both production and
quality control.
HPLC
High-performance liquid chromatography.
IgG
Immunoglobulin G.
IgM
Immunoglobulin M.
Immunization process
A process by which an animal is injected with venom(s) to produce a long-lasting and high-titre
antibody response against the lethal and other deleterious components in the immunogen.
Immunoglobulin
Antibody molecule generated by immunizing an animal (most often a horse) against a snake
venom or a snake venom mixture. Immunoglobulin G (IgG) is the most abundant
immunoglobulin fraction.
In-process control
Checks performed during production to monitor and, if necessary, to adjust the process to ensure
that the antivenom conforms to specifications. The control of the environment or equipment may
also be regarded as part of in-process control.
Manufacture
All operations of purchase of materials and products, production, quality control, release, storage
and distribution of snake antivenom immunoglobulins, and the related controls.
Median effective dose (or effective dose 50%) (ED50)
The quantity of antivenom that protects 50% of test animals injected with a number of LD50 of
venom.
Median lethal dose or lethal dose 50% (LD50)
The quantity of snake venoms, injected intravenously or intraperitoneally, that leads to the death
of 50% of the animals in a group after an established period of time (usually 24–48 hrs).
Milking
The process of collecting venom from live snakes.
4
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Monospecific antivenom
Defines antivenoms that are limited in use to a single species of venomous snake or to a few
closely related species whose venoms show clinically effective cross-neutralization with the
antivenom. The term “monovalent” is often used and has the same meaning.
Mr
Relative molecular mass.
Nanofilter
Filters, most typically with effective pore sizes of 50 nm or below, designed to remove viruses
from protein solutions.
National regulatory authority (NRA)
WHO terminology to refer to national medicines regulatory authorities. Such authorities
promulgate medicine regulations and enforce them.
Plasma
The liquid portion remaining after separation of the cellular elements from blood collected in a
receptacle containing an anticoagulant, or separated by continuous filtration or centrifugation of
anticoagulated blood in an apheresis procedure.
Plasmapheresis
Procedure in which whole blood is removed from the donor, the plasma is separated from the
cellular elements by sedimentation, filtration, or centrifugation, and at least the red blood cells
are returned to the donor.
Polyspecific antivenom
Defines antivenoms that are obtained by fractionating the plasma from animals immunized by a
mixture of venoms from several species of venomous snakes. The term “polyvalent” is often
used and has the same meaning.
Prion
A particle of protein that is thought to be able to self-replicate and to be the agent of infection in
a variety of diseases of the nervous system, such as mad cow disease and other transmissible
spongiform encephalopathies (TSE). It is generally believed not to contain nucleic acid.
Production
All operations involved in the preparation of snake antivenom immunoglobulins, from
preparation of venoms, immunization of animals, collection of blood or plasma, processing,
packaging and labeling, to its completion as a finished product.
Quarantine
A period of enforced isolation and observation typically to contain the spread of an infectious
disease among animals. The same terminology applies to the period of isolation used to perform
quality control of plasma prior to fractionation, or of antivenom immunoglobulins prior to
release and distribution.
5
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
SDS–PAGE
Sodium dodecyl sulfate–polyacrylamide gel electrophoresis.
Serum
A liquid portion remaining after clotting of the blood. Serum has a composition similar to plasma
(including the immunoglobulins) apart from fibrinogen and other coagulation factors which
constitute the fibrin clot.
Standard operating procedure (SOP)
An authorized written procedure giving instructions for performing operations not necessarily
specific to a given product or material (e.g. equipment operation, maintenance and cleaning;
validation; cleaning of premises and environmental control; sampling and inspection). Certain
SOPs may be used to supplement product-specific master and batch production documentation.
Toxin
A toxic substance, especially a protein, which is produced by living cells or organisms and is
capable of causing disease when introduced into the body tissues. It is often also capable of
inducing neutralizing antibodies or antitoxins.
Traceability
Ability to trace each individual snake, venom, immunized animal, or unit of blood or plasma
used in the production of an antivenom immunoglobulin with the final fractionated batch. The
term is used to describe forward and reverse tracing.
TSE
Transmissible spongiform encephalopathy.
Validation
Action of proving, in accordance with the principles of good manufacturing practice, that any
procedure, process, equipment, material, activity, or system actually leads to the expected results.
Venom
The toxic secretion of a specialized venom gland which, in the case of snakes, is delivered
through the fangs and provokes deleterious effects. Venoms usually comprise many different
protein components of variable structure and toxicity.
Viral inactivation
A process of enhancing viral safety in which viruses are intentionally “killed”.
Viral reduction
A process of enhancing viral safety in which viruses are inactivated and/or removed.
Viral removal
A process of enhancing viral safety by partitioning viruses from the protein(s) of interest.
6
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
3
GENERAL CONSIDERATIONS
Snake antivenom immunoglobulins (antivenoms) are the only specific treatment for envenoming
by snakebites. They are produced by the fractionation of plasma usually obtained from large
domestic animals hyper-immunized against relevant venoms. Important but infrequently used
antivenoms may be prepared in small animals. When injected into an envenomed human patient,
antivenom will neutralize any of the venoms used in its production, and in some instances will
also neutralize venoms from closely related species.
3.1
Historical background
Shortly after the identification of diphtheria and tetanus toxins, von Behring and Kitasato
reported the antitoxic properties of the serum of animals immunized against diphtheria or tetanus
toxins and suggested the use of antisera for the treatment of these diseases (2). In 1894, von
Behring diphtheria antitoxin was first successfully administered by Roux to save children
suffering from severe diphtheria. Thus, serum therapy was born and the antitoxin was
manufactured by Burroughs Wellcome in the United Kingdom. The same year, Phisalix and
Bertrand (3) and Calmette (4) simultaneously, but independently, presented during the same
session of the same meeting their observations on the antitoxic properties of the serum of rabbits
and guinea-pigs immunized against cobra and viper venoms, respectively. Immediately after his
discovery of “antivenin serum-therapy”, Albert Calmette was actively involved in proving its
efficacy in the treatment of human envenoming. The first horse-derived antivenom sera that he
prepared were already in clinical use in 1895 by Haffkine in India and by Lépinay in Viet Nam.
The latter reported the first successful use of antivenin serum therapy in patients in 1896 (5).
3.2
The use of serum versus plasma as source material
Historically, the pioneers Calmette, Vital Brazil and others, used serum separated from the blood
of hyperimmunized horses for the preparation of antivenom (“antivenin serum-therapy”). Later,
antibodies (immunoglobulins) were demonstrated to be the active molecules responsible for the
therapeutic action of “antivenom serum”. Subsequently, immunoglobulins, or immunoglobulin
fragments (F(ab')2), purified from serum were used instead of crude serum (6, 7).
Nowadays, plasmapheresis, whereby erythrocytes are re-injected into the donor animal within 24
hours of blood collection, is commonly employed to reduce anaemia in the hyperimmunized
animal that donates the plasma. Accordingly, it is almost exclusively, plasma rather than serum
that is used as the starting material for the extraction of the immunoglobulin or its fragments (8–
10). Thus “snake antivenom immunoglobulin” is the preferred term, rather than “anti-snakebite
serum” or “antiserum” which are no longer accurate.
3.3
Antivenom purification methods and product safety
Purification methods were introduced to reduce the frequency of antivenom reactions by
removing the Fc fragment from IgG, thus preventing complement activation and perhaps
reducing the intensity of immune-complex formation responsible for late antivenom reactions
(serum sickness). For 60–70 years, immunoglobulin F(ab')2 fragments have been widely used.
However, antivenom protein aggregation, and not Fc-mediated complement activation, was
increasingly identified as a major cause of antivenom reactions. Thus, a critical issue in
antivenom safety probably lies in the physicochemical characteristics of antivenoms and not
exclusively in the type of neutralizing molecules constituting the active substance. It is also
important to ensure that the current methodologies to produce antivenoms provide a sufficient
margin of safety with regard to the risk of transmission of zoonosis.
7
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
3.4
Pharmacokinetics and pharmacodynamics of antivenoms
Rapid elimination of some therapeutic antivenoms (e.g. when Fab fragments are used) has led to
recurrence of envenoming in patients. However, the choice of preparing specific IgG or
fragments appears to depend on the size and toxicokinetics of the principal toxin(s) of the
venoms. Large relative molecular mass (Mr) bivalent antibodies (IgG and F(ab')2 fragments) may
be effective for the complete and prolonged neutralization of intravascular toxins (e.g.
procoagulant enzymes) which have a long half-life in envenomed patients, whereas low Mr and
monovalent IgG fragments such as Fab may be more appropriate against low-molecular-mass
neurotoxins which are rapidly distributed to their tissue targets and are rapidly eliminated from
the patient’s body (11).
3.5
Need for national and regional reference venom preparations
Antivenom production is technically demanding. The need to design appropriate polyspecific
antivenoms is supported by the difference in venom composition among venomous animals,
associated with the fact that:
— many countries can be inhabited by several medically important species; and
— in many circumstances there is no distinctive clinical syndrome to direct the use of
monospecific antivenoms.
However, similarities in the venom toxins of closely related venomous species may result in
cross-neutralization, thus reducing the number of venoms required for the preparation of
polyspecific antivenoms. Cross-neutralization should be tested in animal models and ideally by
clinical studies in envenomed patients. Preclinical testing of antivenoms against medically
important venoms present in each geographical region or country is a prerequisite for product
licences and batch approval, and should always precede clinical use in envenomed patients. This
requires efforts by manufacturers and/or regulators to establish regional or national reference
venom preparations that can be used to test the neutralization capacity of antivenoms. The
national control laboratory of the country where the antivenom will be used, or the manufacturer
seeking a licence for the antivenom, should perform such preclinical testing using reference
venom preparations relevant to the country or the geographical area.
4
EPIDEMIOLOGICAL BACKGROUND
The incidence of snakebites in different parts of the world and the recognition of the particular
species of greatest medical importance is fundamental to the appropriate design of monospecific
and polyspecific antivenoms in countries and regions. Up-to-date knowledge is therefore highly
relevant to antivenom manufacturers and regulators, especially for the selection of the most
appropriate venoms, or venom mixtures, to be used in the production and quality control of
antivenoms.
4.1
Global burden of snakebites
Envenoming and deaths resulting from snakebites are a particularly important public health
problem in rural tropical areas of Africa, Asia, Latin America and New Guinea (12). Agricultural
workers and children are the most affected groups. Epidemiological assessment of the true
incidence of global mortality and morbidity from snakebite envenomings has been hindered by
several well recognized problems (13, 14). Snakebite envenomings and associated mortality are
under-reported because many victims (20–70% in some studies) do not seek treatment in
government dispensaries or hospitals and hence are not recorded. This occurs because medical
8
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
posts in regions of high incidence are unable to keep accurate records and because death
certification of snakebite is often imprecise (15, 16). Correctly designed population surveys, in
which questionnaires are distributed to randomly selected households in demographically welldefined areas, are the only reliable method for estimating the true burden of snakebites in rural
areas. The results of the few such surveys that have been performed have shown surprisingly
high rates of bites, deaths and permanent sequelae of envenoming (15,
17–20). However, because of the heterogeneity of snakebite incidence within countries, the
results of surveys of local areas cannot be extrapolated to give total national values. Most of the
available data suffer from these deficiencies and, in general, should be regarded as
underestimates and approximations. Published estimates of global burden suggest a range from a
minimum of 421 000 envenomings and 20 000 deaths up to as high as 2.5 million cases and over
100 000 deaths each year (14, 21). In addition, the number of people left with permanent
sequelae as a result of these envenomings is likely to be higher than the number of fatalities (12).
As already identified, most of the estimated burden of snakebite is from sub-Saharan Africa,
south and south-east Asia and central and south America.
The current literature on snakebite epidemiology highlights the inadequacy of the available data
on this neglected tropical disease. There is clearly a need to improve reporting and recordkeeping of venomous bites in health facilities, to support high-quality epidemiological studies of
snakebite in different regions, and to improve the training of medical personnel. Wherever
possible, recording the species that caused the bite as well as death or injury would greatly assist
in documenting which species are of clinical significance in individual countries. Making
venomous bites notifiable and fully implementing the use of the International Statistical
Classification of Diseases and Related Health Problems 10th Revision (22) in official death
certification (e.g. T 63.0 snake venom) would further help to determine the burden of snakebite
more accurately.
4.2
Main recommendations
In most parts of the world, snakebites are under-reported and in some parts are completely
unreported. This deficiency in surveillance and the paucity of properly designed epidemiological
studies explain why the impact of this important public health problem has remained for so long
unrecognized and neglected.
National health authorities should be encouraged to improve the scope and precision of their
epidemiological surveillance of this disease by:
•
improving the training of all medical personnel so that they are more aware of the
local causes, manifestations and treatment of venomous bites;
•
making venomous bites notifiable diseases;
•
setting up standardized and consistent epidemiological surveys;
•
improving the reporting and record keeping of venomous bites by hospitals, clinics,
dispensaries and primary health care posts, relating the bites to the species of
venomous snake that caused the bite wherever possible; and
•
fully implementing the use of the International Statistical Classification of Diseases
and Related Health Problems 10th Revision (2007) (22) in official death
certification (e.g. T 63.0 snake venom)1.
1
http://www.who.int/classifications/apps/icd/icd10online/
9
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
5
5.1
WORLDWIDE DISTRIBUTION OF VENOMOUS SNAKES
Taxonomy of venomous snakes
Recognizing the species causing the greatest public health burden, designing and manufacturing
antivenoms and optimizing patient treatment are all critically dependent on a correct
understanding of the taxonomy of venomous snakes. Like other sciences, the field of taxonomy
is constantly developing. New species are still being discovered, and many species formerly
recognized as being widespread have been found to comprise multiple separate species as
scientists obtain better information, often with new technologies. As the understanding of the
relationships among species is still developing, the classification of species into genera is also
subject to change. The names of venomous species used in these guidelines conform to the
taxonomic nomenclature that was current at the time of preparation. Some groups of venomous
snakes remain under-studied and poorly known. In these cases, the classification best supported
by what evidence exists is presented with the limitation that new studies may result in changes to
the nomenclature.
Clinicians, toxinologists, venom producers and antivenom manufacturers should endeavour to
remain abreast of these nomenclatural changes. These changes often reflect improved knowledge
of the heterogeneity of snake populations, and may have implications for venom producers,
researchers and antivenom manufacturers. Although taxonomic changes do not necessarily
indicate the presence of “new” venoms, they strongly suggest that toxinological and
epidemiological research into these “new” taxa may be required to establish their medical
relevance, if any.
Since some of the names of medically important species have changed in recent years, the
following points are intended to enable readers to relate the current nomenclature to information
in the former literature.
− The large group of Asian pit vipers, which for many years were referred to as a single genus
(Trimeresurus), have been split into a number of new genera (e.g. Cryptelytrops, Parias,
Peltopelor, Himalayophis, Popeia, Viridovipera, Ovophis and Protobothrops, with a few
species retained in Trimeresurus) based on current views of the inter-relationships between
these groups. There are divergent views on this approach to the taxonomy of these snakes,
and interested parties should consult the literature. Some changes which occurred in the early
1980s have only gained wider acceptance during the past decade (i.e. Protobothrops).
Medically important species formerly classified in Trimeresurus include Cryptelytrops
albolabris, C. erythrurus, C. insularis, Protobothrops flavoviridis, P. mucrosquamatus and
Viridovipera stejnegeri.
− It is likely that new species of cobra (Naja spp.) will be identified within existing taxa in both
Africa and Asia; three new species (N. ashei, N. mandalayensis and N. nubiae) have been
described and several subspecies elevated to specific status since 2000 (e.g. Naja annulifera
and N. anchietae, from being subspecies of N. haje), in addition to the recent synonymization
of the genera Boulengerina and Paranaja within the Naja genus. Such changes may hold
significance for antivenom manufacturers and should stimulate further research to test
whether existing antivenoms cover all target snake populations.
− Several medically important vipers have been reclassified: Daboia siamensis has been
recognized as a separate species from Daboia russelii; Macrovipera mauritanica and M.
deserti have been transferred to Daboia; the Central American rattlesnakes, formerly
classified with Crotalus durissus, are now Crotalus simus; and Bothrops neuwiedi has been
found to consist of a number of different species, three of which (B. neuwiedi, B. diporus and
B. mattogrossensis) are of public health importance.
10
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
It is recognized that there have been many accepted revisions of taxonomy over the past few
decades. These Guidelines are aimed at a very wide range of readers, and to assist users in
matching some old and familiar names with the current nomenclature, Tables 1 and 2 summarize
major changes between 1999 and 2008. Users are also encouraged to refer to appropriate
references listed in the document.
Table 1
Genus-level name changes (1999–2008)
Currently accepted name
Previous name/s
Bothrocophias hyoprora
Bothrocophias microphthalmus
Cryptelytrops albolabris
Cryptelytrops erythrurus
Bothrops hyoprora
Bothrops microphthalmus
Trimeresurus albolabris
Trimeresurus erythrurus
Trimeresurus insularis,
Trimeresurus albolabris insularis
Trimeresurus macrops
Trimeresurus purpureomaculatus
Trimeresurus septentrionalis,
Trimeresurus albolabris septentrionalis
Macrovipera deserti, Vipera mauritanica deserti,
Vipera lebetina deserti
Macrovipera mauritanica,
Vipera lebetina mauritanica
Vipera palaestinae
Vipera russelii
Trimeresurus tibetanus
Vipera raddei
Vipera xanthina
Boulengerina annulata
Boulengerina christyi
Trimeresurus flavomaculatus
Trimeresurus sumatranus
Zhaoermia mangshanensis, Ermia mangshanensis,
Trimeresurus mangshanensis
Trimeresurus stejnegeri
Cryptelytrops insularis
Cryptelytrops macrops
Cryptelytrops purpureomaculatus
Cryptelytrops septentrionalis
Daboia deserti
Daboia mauritanica
Daboia palaestinae
Daboia russelii
Himalayophis tibetanus
Montivipera raddei
Montivipera xanthina
Naja annulata
Naja christyi
Parias flavomaculatus
Parias sumatranus
Protobothrops mangshanensis
Viridovipera stejnegeri
11
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Table 2
Changes resulting from new species descriptions, or species redefinitions (1999–2008)
Currently accepted name
Acanthophis laevis (New Guinea)
Acanthophis rugosus (New Guinea)
Agkistrodon taylori
Bitis gabonica
Bitis rhinoceros
Bothrops diporus
Bothrops mattogrossensis
Bothrops pubescens
Crotalus oreganus
Crotalus simus
Crotalus totonacus
Crotalus tzabcan
Daboia russelii
Daboia siamensis
Echis borkini
Echis omanensis
Gloydius intermedius
Lachesis acrochorda
Naja arabica
Naja anchietae
Naja ashei
Naja nigricincta
Naja nubiae
Naja senegalensis
Pseudechis rossignolii
Pseudonaja aspidorhyncha
Pseudonaja mengdeni
Thelotornis mossambicanus
Thelotornis usambaricus
Tropidolaemus philippensis
Tropidolaemus subannulatus
Walterinnesia morgani
12
Previous name(s)
Acanthophis antarcticus laevis, often confused with A.
antarcticus or A. praelongus but neither occur in New
Guinea
Acanthophis antarcticus rugosus, often confused with A.
antarcticus or A. praelongus but neither occur in New
Guinea
Agkistrodon bilineatus taylori
Bitis gabonica gabonica
Bitis gabonica rhinoceros
Bothrops neuwiedi diporus
Bothrops neuwiedi mattogrossensis, B.n. bolivianus
Bothrops neuwiedi pubescens
Previously considered part of Crotalus viridis
Crotalus durissus durissus (Central American populations
of C. durissus complex)
Crotalus durissus totonacus
Crotalus simus tzabcan, Crotalus durissus tzabcan
Daboia russelii russelii, Daboia r. pulchella
Daboia russelii siamensis, D.r. limitis, D.r. sublimitis,
D.r. formosensis
Previously considered part of Echis pyramidum
Previously considered as north-eastern population of Echis
coloratus
Previously named Gloydius saxatilis
Previously considered part of Lachesis stenophrys
Previously considered part of Naja haje
Naja annulifera anchietae, Naja haje anchietae
Previously considered part of Naja nigricollis
Naja nigricollis nigricincta, Naja nigricollis woodi
Previously considered part of Naja pallida
Previously considered part of Naja haje
First described as Pailsus rossignolii, previously considered
part of Pseudechis australis
Previously considered part of Pseudonaja nuchalis
Previously considered part of Pseudonaja nuchalis
Thelotornis capensis mossambicanus
Thelotornis capensis mossambicanus
Previously considered part of Tropidolaemus wagleri
Previously considered part of Tropidolaemus wagleri
Previously considered part of Walterinnesia aegyptia
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
5.2
Medically important venomous snakes
Based on current herpetological and medical literature, it is possible to partially prioritize the
species of snakes that are of greatest medical importance in different regions. Detailed statistics
on the species of snakes responsible for envenomings and fatalities throughout the world are
lacking, except for a few epidemiological studies which include rigorous identification of the
biting snake in a few scattered localities. Thus, establishing a list of medically important species
for different countries, territories and other areas relies, at least in part, on extrapolation from the
few known studies, as well as on the biology of the snake species concerned: e.g. where species
of a group of snakes are known to be of public health importance, based on epidemiological
studies, it seems reasonable to deduce that closely related species with similar natural history
occurring in hitherto unstudied regions are also likely to be medically important. Examples
include Asian cobras in several under-studied regions of Asia, lowland Bungarus species in Asia,
and spitting cobras in Africa.
Tables 3–6 list the species of venomous snakes of greatest medical importance in each of four
broad geographical regions. Species listed in these tables are either:
— those which are common or widespread in areas with large human populations and which
cause numerous snakebites, resulting in high levels of morbidity, disability or mortality
among victims; or
— poorly known species that are strongly suspected of falling into this category; or
— species which cause major and life-threatening envenoming responsive to antivenom, but are
not common causes of bites.
The venoms of these species should be considered a starting point for establishing the most
important targets for antivenom production. The need for additional epidemiological and
toxinological research to better define which venoms to include and exclude for antivenom
production in various regions, territories and countries around the world is emphasized.
Detailed data regarding countries, territories and other areas on species believed to contribute to
the global burden of injury and/or to pose significant risk of morbidity or mortality are provided
in Appendix 1.
13
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Table 3
Venomous snakes of highest medical importance: Africa and the Middle East
North Africa/Middle East
Atractaspididae: Atractaspis andersonii; Elapidae: Naja arabica, Naja haje, Naja oxiana;
Viperidae: Bitis arietans; Cerastes cerastes, Cerastes gasperettii; Daboia mauritanica1, Daboia
palaestinae1; Echis borkini, Echis carinatus, Echis coloratus, Echis omanensis1, Echis
pyramidum; Macrovipera lebetina, Montivipera xanthina1; Pseudocerastes persicus
Central sub-Saharan Africa
Elapidae: Dendroaspis jamesoni, Dendroaspis polylepis; Naja anchietae1, Naja haje, Naja
melanoleuca, Naja nigricollis; Viperidae: Bitis arietans, Bitis gabonica1, Bitis nasicornis; Echis
leucogaster, Echis ocellatus, Echis pyramidum
Eastern sub-Saharan Africa
Elapidae: Dendroaspis angusticeps, Dendroaspis jamesoni, Dendroaspis polylepis; Naja
anchietae1, Naja annulifera, Naja ashei1, Naja haje, Naja melanoleuca, Naja mossambica, Naja
nigricollis; Viperidae: Bitis arietans, Bitis gabonica1, Bitis nasicornis; Echis pyramidum
Southern sub-Saharan Africa
Elapidae: Dendroaspis angusticeps, Dendroaspis polylepis; Naja anchietae1, Naja annulifera,
Naja mossambica, Naja nigricincta1, Naja nivea; Viperidae: Bitis arietans
Western sub-Saharan Africa
Elapidae: Dendroaspis jamesoni, Dendroaspis polylepis, Dendroaspis viridis; Naja haje, Naja
katiensis, Naja melanoleuca, Naja nigricollis, Naja senegalensis; Viperidae: Bitis arietans, Bitis
gabonica1, Bitis nasicornis, Bitis rhinoceros1; Cerastes cerastes; Echis jogeri, Echis leucogaster,
Echis ocellatus
Table 4
Venomous snakes of highest medical importance: Asia and Australasia
Central Asia
Elapidae: Naja oxiana; Viperidae: Echis carinatus; Gloydius halys; Macrovipera lebetina
East Asia
Elapidae: Bungarus multicinctus; Naja atra; Viperidae: Cryptelytrops albolabris1; Daboia
siamensis1; Deinagkistrodon acutus; Gloydius blomhoffii, Gloydius brevicaudus; Protobothrops
flavoviridis, Protobothrops mucrosquamatus; Viridovipera stejnegeri1
South Asia
Elapidae: Bungarus caeruleus, Bungarus niger, Bungarus sindanus, Bungarus walli; Naja
kaouthia, Naja naja, Naja oxiana; Viperidae: Cryptelytrops erythrurus1; Daboia russelii1; Echis
carinatus; Hypnale hypnale; Macrovipera lebetina
South-East Asia (excluding Indonesian West Papua)
Elapidae: Bungarus candidus, Bungarus magnimaculatus, Bungarus multicinctus, Naja atra,
Naja kaouthia, Naja mandalayensis, Naja philippinensis, Naja samarensis, Naja siamensis, Naja
sputatrix, Naja sumatrana; Viperidae: Calloselasma rhodostoma; Cryptelytrops albolabris1,
Cryptelytrops erythrurus1, Cryptelytrops insularis1; Daboia siamensis1; Deinagkistrodon acutus
Australo-Papua (includes Indonesian West Papua)
Elapidae: Acanthophis laevis1; Notechis scutatus; Oxyuranus scutellatus; Pseudechis australis2,
Pseudonaja affinis, Pseudonaja mengdeni, Pseudonaja nuchalis, Pseudonaja textilis
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
Pseudechis australis is common and widespread and causes numerous snakebites; bites may be severe,
although this species has not caused a death in Australia since 1968.
2
14
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Table 5
Venomous snakes of highest medical importance: Europe
Central Europe
Viperidae: Vipera ammodytes
Eastern Europe
Viperidae: Vipera berus
Western Europe
Viperidae: Vipera aspis, Vipera berus
Table 6
Venomous snakes of highest medical importance: the Americas
North America
Viperidae: Agkistrodon bilineatus, Agkistrodon contortrix, Agkistrodon piscivorus, Agkistrodon
taylori1; Bothrops asper, Crotalus adamanteus, Crotalus atrox, Crotalus horridus, Crotalus
oreganus1, Crotalus simus1, Crotalus scutulatus, Crotalus totonacus1, Crotalus viridis
Caribbean
Viperidae: Bothrops cf. atrox (Trinidad), Bothrops caribbaeus (St Lucia), Bothrops lanceolatus
(Martinique); Crotalus durissus (Aruba)
Central America
Viperidae: Bothrops asper; Crotalus simus1
South America
Viperidae: Bothrops alternatus, Bothrops asper, Bothrops atrox, Bothrops bilineatus, Bothrops
brazili, Bothrops diporus1, Bothrops jararaca, Bothrops jararacussu, Bothrops leucurus,
Bothrops mattogrossensis1, Bothrops moojeni, Bothrops pictus, Bothrops venezuelensis; Crotalus
durissus; Lachesis muta
5.3
Minor venomous snake species
In many countries, territories and other areas there are species of snakes that rarely bite humans
but are capable of causing severe or fatal envenoming. Their medical importance may not justify
inclusion of their venoms in the immunizing mixture for production of polyspecific antivenoms
but the need to make antivenoms against these species needs to be carefully analysed.
In some cases, such as with some Central American pit vipers (genera Agkistrodon, Porthidium,
Bothriechis, Atropoides among others), there is clinically effective cross-neutralization of
venoms by standard national polyspecific antivenoms (23).
In other cases, there is no effective cross-neutralization and manufacturers may therefore
consider that the production of a small volume of monospecific antivenom is justified for use in
the rare but potentially fatal cases of envenoming, provided that such cases can be identified.
Such antivenoms are currently available for envenoming by the boomslang (Dispholidus typus),
desert black snake (Walterinnesia aegyptia), Arabian burrowing asp (Atractaspis andersonii)
(24), king cobra (Ophiophagus hannah), Malayan krait (Bungarus candidus) (25)
“yamakagashi” (Rhabdophis tigrinus) and red-necked keelback (R. subminiatus), Martinique’s
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
15
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
“Fer-de-lance” (Bothrops lanceolatus), St Lucia’s B. caribbaeus, and some species of American
coral snake (Micrurus).
No antivenoms are currently available for envenoming by species such as African bush vipers
(e.g. Atheris, Proatheris), berg adder (Bitis atropos) and several other small southern African
Bitis spp. (e.g. B. peringueyi), Sri Lankan and south-west Indian humpnosed vipers (Hypnale
spp.) (26, 27), many Asian pit vipers (“Trimeresurus” sensu lato), some species of kraits
(e.g. B. niger) and all but one species of burrowing asp (genus Atractaspis).
An alternative to antivenom production against species that cause few, but potentially severe
accidents, is to manufacture polyspecific antivenoms for broadly distributed groups that have
similar venom compositions (e.g. African Dendroaspis and Atractaspis; Asian “green pit
vipers”; American Micrurus). This may result in antivenoms that offer broad protection against
venoms from minor species within genera, or species whose bites are less frequent than those of
others in the same taxonomic groups (i.e. genus, sub-family or family).
5.4
Sea snake venoms
Although venomous marine sea snakes have not been included in the tables of medically
important venomous snakes, it should be recognized that there are a number of species of marine
snakes with potent venoms that can cause illness or death. Available evidence, particularly
clinical experience, indicates that the major sea snake antivenom that is currently commercially
available, which uses venom of a single sea snake, Enhydrina schistosa, plus a terrestrial Elapid,
Notechis scutatus, in the immunizing venoms mixture, is effective against envenomings by other
sea snakes on which there are clinical data. Further research would be needed to better define the
full extent of cross-neutralization offered by this antivenom against other sea snake species.
5.5
Main recommendations
•
Clinicians, toxinologists, poison centres, regulators, venom producers and antivenom
manufacturers should be well-informed about current nomenclature and new changes
to taxonomy, so as to ensure the currency of information, correct identification of
species in their countries, and correct selection and sourcing of venoms used in the
manufacture of antivenoms.
•
Identification of the medically important venomous snakes that cause the greatest
burden of injury, disability and/or mortality is a critical prerequisite to meeting the
need for efficacious antivenom. Improving the quality of the available data and
correcting and amplifying the level of geographical detail and precision of attribution
should be important priorities.
•
Support for establishment of local capacity for venom production as a means of
ensuring that venom immunogens from geographically representative populations of
medically important snake species are used in antivenom production should improve
antivenom specificity.
16
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
6
ANTIVENOMS DESIGN: SELECTION OF SNAKE VENOMS
An accurate selection of snake venoms is critical for the production of antivenoms that have the
capacity to cover the majority of cases of envenoming in a given geographical region, territory or
country. The composition of snake venoms is very complex and a high inter-species and intraspecies variation has been documented (28). Therefore the design of the antigenic mixture to be
used in antivenom production is a critical task.
The selection of the most appropriate snake venoms for the production of antivenoms needs to be
carefully analysed and should take into consideration:
— the geographical region where the antivenom is going to be used;
— the medically most relevant snakes from the geographical region where the antivenom is
going to be used;
— the variability of venom composition within the region of distribution of a snake species;
— the information on cross-neutralization of antivenoms against the venoms of species not
included in the mixture of venoms used to immunize animals for antivenom manufacture.
6.1
Selection and preparation of representative venom mixtures
Appendix 1 presents an up-to-date list of the most relevant species of snakes, from a medical
standpoint, in the various regions, countries and territories of the world. Manufacturers should
consider, as a priority, the venoms of species included in category 1 of this Appendix for the
design of venom mixtures for immunization. Venoms to include in a venom pool used for animal
immunization should be selected on the basis of the geographical region where an antivenom is
intended to be distributed. On a case by case basis, venoms from species listed in category 2
could be included in an immunizing mixture.
There are variations in venom composition and antigenicity within the geographical range of a
single taxonomic species as well as other causes of intra-species variation (such as changes
according to the age of the specimens) (29, 30). Therefore, pooled representative samples of
venoms should be prepared from snakes of different geographical origins and ages (see section 7
on venom preparation). Cross-neutralization of venoms outside the range of venoms used for
immunization may extend the range of therapeutic applications of some antivenoms. Results of
preclinical potency testing may be used to identify a potential cross-neutralization capacity of
antivenoms, which should subsequently be confirmed by clinical testing in envenomed patients.
In vitro immunological cross-reactivity should not be used as the single basis for recommending
therapeutic use of an antivenom outside the range of venoms used in its production.
6.2
Manufacture of monospecific or polyspecific antivenoms
A major issue in designing antivenoms is to define whether they should have monospecific or
polyspecific activity.
6.2.1 Monospecific antivenoms
Monospecific antivenoms are limited in use to a single species of venomous snake or to a few
closely related species whose venoms show clinically effective cross-neutralization. These
conditions apply in areas where:
− there is only one medically important species (e.g. Vipera berus in the United Kingdom
and Scandinavia);
17
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
− a simple blood test, suitable for use even in peripheral health care centres, can define the
biting species (e.g. detection of incoagulable blood by the 20-minute whole blood
clotting test in the northern third of Africa where only Echis spp. cause coagulopathy);
− a simple algorithmic approach allows the species to be inferred from the pattern of
clinical and biological features;
− there is a reliable and affordable rapid immunodiagnostic test readily available allowing
the toxins to be identified unambiguously.
However, most countries are inhabited by several medically important species of snakes, where
there may be no distinctive clinical syndrome to direct the use of a monospecific antivenom. In
these cases, the manufacture of polyspecific antivenoms should be highly recommended.
6.2.2 Polyspecific antivenoms
Some clinicians are uncertain about using polyspecific antivenoms because of a fear that they
have an inherently lower potency than monospecific antivenoms but this is not necessarily the
case.
Polyspecific antivenoms can be generated by immunizing animals with a mixture of venoms
from various snake species. The resulting antivenom will then contain antibodies against venom
components of various snake species. When a polyspecific antivenom is produced this way, by
immunizing an animal with venoms from several taxonomically-related snakes (e.g. different
vipers), the titre of neutralizing antibodies against individual venoms may in some cases be
higher than in a monospecific antivenom produced by immunizing an animal with only a single
venom (31).
Polyspecific antivenoms can also be obtained either by:
— immunizing individual animals with the venom of a single species and then mixing the
various hyperimmune plasmas for fractionation; or
— mixing appropriate quantities of the relevant purified antivenoms before formulation.
When using the latter option it is important to monitor the potency for each monospecific
antivenom to ensure that the potency of the mix in the final product is consistent, reproducible
and in line with the product specification for each individual venom. A sufficiently high titre
should be guaranteed to avoid the need for infusion of an additional antivenom dose to the
patient, as this may increase the risks of adverse reactions. However, in such “combined
antivenoms”, neutralizing antibodies against all individual venoms will be proportionally diluted.
In general, such dilution implies that a greater amount of antivenom proteins would be infused to
patients, which is likely to increase the risks of adverse reactions.
In some regions, it is possible to differentiate envenomings on the basis of obvious distinct
clinical effects: neurotoxicity, local tissue damage and/or haematological disturbances
(haemorrhage or coagulopathy). Such situations justify the preparation of separate polyspecific
antivenoms against mixtures of either neurotoxic venoms or venoms inflicting tissue damage,
haemorrhage and/or coagulopathy.
Polyspecific antivenoms offer significant clinical advantages and their production should be
encouraged, whenever technically possible. They can be produced using venoms from a range of
species of venomous snakes of high medical relevance, broadening their usefulness and making
identification of the biting species less critical, having the additional advantage of simplicity of
distribution and supply.
18
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
6.3
7
Main recommendations
•
When selecting snake antivenoms national health authorities should first obtain and
consider the information on the local species and their relative medical importance.
•
The design of the venom mixture used in immunization, and the decision to prepare
monospecific or polyspecific antivenoms, depend on the epidemiological and clinical
information on snakebites in that particular region, country or area.
•
Owing to the difficulty of identifying clinically the snake species responsible for
envenoming, polyspecific antivenoms appropriate to the geographical region may be
more practical and convenient to use than monospecific antivenoms.
•
Polyspecific antivenom may be prepared either by mixing monospecific antivenoms
or by immunizing animals with a mixture of venoms, provided that the specified
neutralizing titre for each venom is achieved. The preparation of antivenoms by
immunizing animals with a mixture of venoms from taxonomically related snakes
may result in a higher titre antivenom.
•
Manufacturers seeking marketing authorization for antivenoms in a given country
should provide experimental evidence from preclinical testing that the product
exhibits a neutralization capacity against different local venoms (see section 17).
PREPARATION AND STORAGE OF SNAKE VENOM
Venom preparations are used both to hyper-immunize animals, as part of antivenom production,
and to provide reference venom samples for routine and/or preclinical potency assessment of
antivenoms. Ensuring their quality is therefore critical, and their preparation should follow the
principles and recommendations mentioned below. The essential principles of quality systems
should be followed in venom production including traceability, reproducibility, taxonomic
accuracy, and hygiene control.
Venoms used for antivenom manufacture should be representative of the snake population living
in the area where the antivenom is to be used. To take account of the variability in venom
composition of an individual species (32–34), it is imperative that the venom of an adequate
number of snakes (generally not less than 20–50 specimens) from the same geographical location
should be collected together. A similar preparation can be used as a national standard of venoms
for routine potency assessment of antivenoms (see section 8) and to perform preclinical testing
of antivenoms (see section 17) to verify that the antivenom is suitable to treat envenomings in
the region efficiently. Venom producers should follow rigorously the recommendations listed
below and provide evidence of compliance on:
— geographical origin and size (and hence the approximate age) of each individual snake used
for venom production;
— taxonomic details of each snake used;
— correct implementation of the Convention on International Trade in Endangered Species
(CITES) documents in the case of endangered species;
— precautionary measures to avoid collection of venoms from sick snakes;
— individual identification of snakes contributing to each venom batch; and
— traceability of each venom batch.
19
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Being able to fulfil the following is also strongly recommended:
— rapid freezing of the venom after collection;
— lyophilization of the venom for long-term storage1; and
— confirmation of batch-to-batch similarity of venom of the same origin.
7.1
Production of snake venoms for immunization
The maintenance of a snake farm and the handling of snakes used for antivenom production
should comply with quality systems principles.
7.1.1 Quarantine of snakes
All new accessions should be quarantined for at least 2 months in a special room (“quarantine
room”) which should be located as far as possible from the “production rooms” where snakes
qualified for milking are kept.
On arrival, snakes should be examined by a specialized veterinary surgeon (or experienced
person) for ectoparasites and pentastomids – which should be eliminated using broad-spectrum
antiparasitic drugs – and possible infections, particularly transmissible ones (35–37). Some
viruses can be transmitted between different species (for example from Bothrops spp. to
Crotalus spp.).
Sick snakes should be treated and their quarantine extended for 2 months after complete clinical
recovery. Sick animals found in “production rooms” may be treated in situ but they cannot be
milked for venom production. If an antibiotic treatment is given, the snake should not be milked
for 4 weeks following the end of the treatment. When housed in good conditions, adult snakes
collected from the wild can live on a snake farm for 10 years or more. When handling snakes,
the risk of infection with human mosquito-borne viruses such as Japanese encephalitis should be
prevented, since arbovirus infections have been reported in some snakes (38).
7.1.2 Maintenance of captive snakes for venom production
Individual snakes should preferably be housed in separate cages large enough to allow them to
move about. There are several acceptable options for the design of the cages. Transparent or
black (for burrowing snakes) plastic boxes are recommended. Cage materials should be
impermeable, free from fissures, and inert to disinfectants, cleaning chemicals and common
solvents. The selection of cleaning and disinfecting agents should be carefully considered to
ensure they do not have adverse effects on the snakes. Cages should be adequately ventilated but
perforations or mesh should be small enough to prevent escape. In the case of gravid female
vipers, the mesh should be sufficiently fine to prevent escape of their tiny, live-born babies. The
cage interior should be visible from the outside to allow safe maintenance and handling. Access
to cages through doors, lids or sliding panels should facilitate management without
compromising safety or allowing snakes to escape. A disposable floor covering (e.g. newspaper)
is recommended. Cryptic and nocturnal species should be provided with a small shelter where
they can hide.
The use of “hide boxes” is increasingly common as these provide both a more reassuring
environment for the snake, and increased safety for keepers. Hide boxes should be designed to be
slightly larger than the curled snake, with a single small entrance/exit hole, large enough to allow
a recently fed snake easy access, plus some simple closure device to lock the snake in the hide
1
20
Desiccation may be acceptable if proven to ensure stability of the preparation.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
box. This will allow removal of the snake from the cage without hazard to the keeper, making
routine cage maintenance simpler and safer. Hide boxes can be plastic or wooden, but should be
readily cleanable. The roof of the hide box should be removable, to allow easy, safe extraction of
the snake, when required.
Cages should be thoroughly cleaned and disinfected, ideally when soiled (perhaps almost daily
for elapids), but at least every week. Faeces and uneaten or regurgitated rodents should be
removed. To avoid misidentification of the snake, a label bearing its individual data should be
attached to the cage and transferred with the snake when it is moved to another cage. Water
should be provided on at least two days per week, but for species from humid climates, more
frequent watering or misting may be required, particularly when sloughing. Water should be
changed regularly and as soon as it becomes contaminated. Water treatment by ultraviolet (UV)
sterilization or acidification may be considered.
Tens of cages may be accommodated in the same “production room”, provided that there is
enough space for maintenance and milking. This room should be kept as clean as possible and
thoroughly cleaned at least weekly. Access should be guarded by a tray containing an antiseptic
which is placed on the floor at the entrance so that the footwear of all people entering is
automatically treated. The temperature and humidity of the snake room should be controlled
according to the climatic requirements of the particular snake species. Ventilation should be
ensured using fans, air conditioning, or air renewing systems.
Access to snake rooms should be restricted to personnel responsible for their maintenance. They
should be kept locked, with any windows permanently closed or protected by bars and mosquito
proofing. Access should be via a safety porch not allowing simultaneous door opening and with
a transparent panel allowing a view of the entire snake room to check whether any animals have
escaped from their cages. The spaces below the doors should be less than 3 mm and all openings
to the exterior (e.g. water pipes, drainage conduits, ventilation entrances and exits) should be
protected by grilles having holes smaller than 5 mm. Natural light is often used; however, when
not available, artificial light should be turned on for 12 hours during the day and turned off
during the night for tropical species, but species from temperate zones may have different
requirements. Snakes of the same species, collected at the same time in the same area should be
placed in the same racks. The same “production room” can contain snakes of different species,
provided that they have similar living requirements (i.e. temperature and humidity).
When kept under favourable housing and climatic conditions, and if left undisturbed, snakes will
reproduce in captivity (39). Animals should be mated only with specimens from the same
species, subspecies and local origin (40, 41). Sexing can be difficult, but is helped by the use of
intra-cloacal probes. The male and the female should be individually identified and separated
soon after copulation. The female should be kept under careful surveillance. Eggs from
oviparous snakes and newborns from ovoviviparous snakes should be removed from their
mother’s cage as soon as possible. When a difference in the venom composition of adult and
juvenile snakes has been reported, as in the case of Bothrops and Crotalus species (32, 42–44),
the venom of a certain proportion of juvenile snakes might be mixed with that of adults.
The ideal frequency of feeding captive snakes depends on the species and age of the snake,
varying from twice per week to once per month. Snakes are usually fed after being milked,
ideally with dead mice or other appropriate prey according to the snake species. Some snakes
will only accept living prey, but attempts should be made to wean them onto dead prey. Snakeeating species, such as kraits, coral snakes and king cobras, can be enticed to take dead mice if
the prey is first flavoured with snake tissue fluids or even snake faeces. Living, dead or
regurgitated prey should not be left in the cage for more than a few hours. Force-feeding may be
necessary for neonates and snakes that persistently refuse to feed. Feeding time affords an
21
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
opportunity to carefully check the snake for abnormal behaviour, wounds, and possible
infections and to give dietary supplements when necessary. Individual feeding records are
crucial. They should include details of when prey was offered, when it was consumed and
whether it was regurgitated. The health of captive snakes can be estimated and recorded by
observing regular feeding and by measuring their weight and length. These data are best stored
on a computer system, using a “bar code” for each snake, or, alternatively, using a reliable
manual recording system, and constitute useful records related to the venom batches produced.
Water should be provided in the milking room from a tap, shower or reservoir, as is the case in
laboratories where there is a risk of chemical injuries.
7.1.3 General maintenance of a snake farm
In addition to the rooms devoted to snake housing, sufficient space should be made available for
the storage of consumables, rooms for cleaning and sanitizing cages and racks, animal houses for
rat and mouse production, a storage room for conservation of the venom produced, control
laboratories and administrative offices.
The cage cleaning rooms should be large enough to accommodate all the cages that are being
cleaned and sanitized. Furthermore it is desirable to have two sets of washing and sanitizing
rooms, a larger one for equipment from the venom production room and a smaller one for
equipment from the quarantine area. These rooms should be secure in case a snake, inadvertently
left in its cage, attempts to escape. The cleaning procedures for production rooms and for cages
in which snakes are kept, and the cleaning schedule, should be established and documented.
Food animals, usually rodents, should be purpose-bred in clean conventional animal houses, and
kept, handled and sacrificed in accordance with ethical principles. The rooms for rodent
production should be large enough to provide sufficient numbers of rats or mice to feed the
snakes. Alternatively, rodents can be purchased from qualified commercial sources. Breeding of
rats and mice cannot take place in the same room, because of the stress induced by the rats in the
mice. If snake are bred on the farm, egg incubators, and special rooms for newborns and
juveniles are required. It should be taken into account that the diets required by young snakes
might differ from those of adults (for instance, frogs and tadpoles are preferred to rodents by
some species).
When possible, it is useful to have a small laboratory for performing quality control on the
venoms (see section 8). An area for repairing broken equipment and for other miscellaneous
purposes is also required. The administrative area should be sufficiently large and adequately
equipped with computer facilities, so that the traceability requirements needed for venom
production can be met. The whole venom production facility should be made secure against
unauthorized intrusion.
7.1.4 Snake milking for venom production
Specific safety consideration for operators should be applied to snake milking (see section 7.2).
All operations should be fully described in written procedures, which should be checked and
revised periodically according to a written master document. Pools of venom require unique
batch numbers, and the snake milkings contributing to the pool should be traceable.
7.1.4.1 Venom collection on snake farms
Snakes can be milked according to a regular schedule, depending on the species. The interval
between milkings varies among producers and ranges from every 2 or 3 weeks to every 3
months, except for specimens that are in quarantine or are undergoing treatment and snakes in
the process of sloughing their skins.
22
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Handling equipment must be appropriate for the particular species of snake to cause the least
stress and must be familiar to and afford safety to the operator. The snake is gently removed
from its cage with a hook and either placed on a foam rubber pad before being pinned behind the
head or encouraged to crawl into a transparent plastic tube. For very dangerous species, the use
of short-acting general anaesthesia, or moderate cooling (15 °C) during milking can be
considered (e.g. inhaled sevofluorane or sevoflurane, halothane or even carbon dioxide) as it
reduces the risk of accidents both to the snake and to the snake-handler. Excessive cooling of the
snake in a refrigerator is potentially harmful and is not recommended. For the collection of
venom, the snake’s head is grasped between index finger and thumb, just behind the angle of the
jaw, while the snake’s body is held between the trunk and the arm of the snake handler. An
assistant should gently occlude the snake’s cloaca to prevent messy contamination of the locality
by spraying of faeces.
By applying gentle pressure, the snake's jaws are forced open, the fangs exposed and, in the case
of vipers, erected. In the case of large vipers, the dental sheath is retracted when necessary with
clean forceps. The fangs are pushed through a plastic/parafilm membrane (or the snake may
voluntarily strike through the membrane) hooked over the lip of a glass vessel, and venom is
squeezed out. The use of siliconized containers might be considered to minimize venom
attaching to the container surface. While a brief electrical impulse of moderate intensity can be
applied to stimulate venom secretion, this technique is not used or required by most venom
producers, although it may help in avoiding debris in the venom. Any venom sample
contaminated with blood should be rejected. After venom extraction, the fangs are carefully
withdrawn from the collection vessel, while preventing damage to the mouth and dentition and
avoiding the snake’s impaling itself with its own fangs. After each venom milking, all materials
used for milking should be sterilized with a flame, then cooled with a draught of air before the
next snake is milked.
Special procedures that avoid direct handling should be employed in the case of burrowing asps
(genus Atractaspis) because they cannot be held safely in the way described above (45). In the
case of colubrid snakes, special techniques are required, such as application of foam rubber pads
or capillary tubes to the posteriorly-placed fangs and the use of secretagogue drugs. Similarly,
some elapid snakes have only small fangs capillary tubes or similar are required to collect
venom. At the time of milking, there is an opportunity to remove broken or diseased fangs and to
examine the snake for ectoparasites (e.g. ticks and mites), for pentastomids escaping from the
snake’s respiratory tract and for areas of adherent dead skin and opercular scales over the snake’s
eyes. The snake can be treated with drugs or vitamins at the same time and, if necessary, can be
force-fed. Milking is often combined with cage cleaning and disinfection and the feeding of the
snake. Avoiding trauma to the snake's mouth and dentition is critical to prevent infection and
“mouth rot” and the milking process should be performed following clean practices.
Several snakes from the same group (same species and subspecies collected at the same time in
the same area) can be milked into the same glass vessel. However, it is important for most
venoms to be snap frozen at -20 °C or colder within 1 hour. One method of achieving this for
venoms with high proteolytic activity, is to pour the collected venom into a vial maintained at a
low temperature (ideally at -70 to -80 °C, but, if this is not possible, at -20 to -40 °C), every 10
minutes or at least every 30 minutes, before another snake from the same group is milked.
Centrifugation of freshly collected venom is recommended, for instance at 1000 g for 5 minutes,
since it removes cellular debris.
It is important to identify the vial into which the venom has been collected with an appropriate
reference number. Primary identification must be on the vial. This allows the identification of all
the snakes used, the day of the milking, the name of the operator and any other relevant
information. To obtain large venom batches for the preparation of antivenom, one approach is to
23
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
use the same vial over several months for milking the same snakes, providing the cold chain is
never broken. Pools of venom require unique batch numbers, and the snake milkings
contributing to the pool should be traceable. The venom vial will then be freeze-dried and kept in
the dark at a low temperature (either –20 °C or 4 °C) in a well-sealed flask, precisely identified
with a number, up to the time of delivery. However, some producers use an alternative system,
keeping venom at 20–25 °C in a dessicator. Large pools of frozen venoms, collected from many
individuals, are allowed to thaw at 0 °C, to avoid proteolytic degradation of venom components
and, after being thoroughly mixed, aliquots of liquid venoms are prepared. These aliquots are
then freeze-dried and stored in the dark at a low temperature (either -20 °C or 4 °C). Aliquots of
freeze-dried venoms should be adequately labelled. Freeze-drying cycles should be established,
followed, and documented. Venom stored for considerable periods of time should be tested to
ensure that no degradation or loss of activity has occurred (see section 8).
During milking, the wearing of protective clothing and a mask as well as vinyl gloves is
recommended to prevent any accidents or infections. The equipment used for storage of frozen
venom (freezers) and for freeze-drying, should be cleaned using established procedures, and the
cleaning documented, in order to minimize cross-contamination. Likewise, equipment requiring
calibration, such as freezers, balances and freeze-driers, should be calibrated as per a defined
schedule.
7.1.4.2 Venom collection from wild snakes
In some parts of the world it is accepted practice that during certain seasons, collectors from a
snake farm or local snake catchers will go to designated localities in the wild and collect venom
from snakes manually and release them in the same locality after milking. In such cases,
collecting venoms from wild snakes may be the only alternative, provided the venom and the
collection process are subject to stringent rules, including most of the procedures already
described for captive animals. At any time the collectors may milk from 50 to more than a
hundred snakes; usually these are snakes of smaller size e.g. Echis species.
If collection of venom from wild snakes is necessary, most of the steps and safety procedures
which are followed in milking captive snakes should be adhered to as far as possible, with
certain modifications for the field conditions. The team which goes to the field for collection
should include a herpetologist or zoologist who is able to help and confirm the identity of the
snakes. Sick snakes, injured snakes and gravid females should not be milked. Detailed records of
the locality, season, climate, date, size and number of snakes milked in one batch or pool should
be maintained. One reference specimen from this locality should be taken to the laboratory and
deposited as a voucher specimen for that pool with options for traceability. Where feasible, a
photographic record of snakes milked in the field should be retained.
During milking, the wearing of protective clothing and a mask as well as vinyl gloves is
recommended to prevent any accidents or infections, as for venom collection on a snake farm.
Snake handling and milking should be done in an environment where there is little risk of
external contamination. For example, the milking could be done inside a vehicle rather than in
the open field. The field team should have training in first aid as well as in administration of
antivenom, or they should be within reach of a hospital with antivenom available in case
accidents occur. Milked venom should be frozen as soon as possible in a freezer in the vehicle.
This is particularly important for venoms having high proteolytic activity, such as many viperid
venoms, to avoid enzymatic degradation of venom components.
24
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
7.2
Staff responsible for handling snakes
7.2.1 Safety and health considerations
Handling and milking snakes is a dangerous operation. One envenoming occurred every two
years in each of the 15 extraction facilities reviewed by Powell et al. (46). Twelve bites were
recorded, 10 with envenoming, and one case of venom being squirted into the eye of a worker
(47), between 1981 and 1999, when 370 768 venom extractions were performed at a venom
production plant from Bothrops moojeni.
Milking should be done very carefully by well-trained snake handlers. All personnel involved in
snake handling and venom collection should be fully informed about the potential dangers of
being bitten and envenomed. They should be adequately trained, and the training procedures
must be documented. A minimum of two people should be present during snake handling for
venom collection. For safety reasons, it is recommended that sessions for milking of snakes
should be interrupted at least every 2 hours, for a rest period before re-starting the process.
7.2.2 Clothing for snake and venom handling
Protective clothing should include an eye covering (plastic spectacles), especially when spitting
elapids capable of squirting their venom are being handled, and a laboratory coat or gown. The
wearing of protective gloves designed to prevent an effective bite is unpopular and not usually
recommended because it impairs manual dexterity and sense of touch, but the use of nitrile
gloves is advisable to prevent cross-contamination.
When lyophilized or desiccated venom is being handled, the safety of operators is paramount,
since venom aerosols may form and affect people through skin breaks, eyes or mucous
membranes, or may sensitize them to the venom. Appropriate gowning is necessary when
handling lyophilized venom, to prevent contact of the venom with skin or mucous membranes.
7.2.3 Procedures to be followed if a bite occurs
There are several important measures to be put in place for dealing with a bite (48), as described
below.
7.2.3.1 Procedures and alarms
Clearly defined, prominently displayed, well understood and regularly rehearsed procedures
should be in place in case of a bite. An alarm should be sounded to summon help, the snake
returned safely to its cage or box and the victim should withdraw to an area designated for first
aid.
7.2.3.2 First-aid protocols
Clearly understandable first-aid protocols should be established for each species. These should
be available in printed form adjacent to each cage. Immediate application of pressureimmobilization may be appropriate for treating the bites of rapidly neurotoxic elapids. However,
the technique is not easy and, if they are to use the method properly, staff will need extensive
training and must be provided with the necessary materials (a number of crepe bandages, 10 cm
wide × 4.5 m long, and splints). Provision of appropriate analgesia for first aid should be
considered. If venom enters the eyes, immediate irrigation with generous volumes of clean water
is an urgent necessity.
25
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
7.2.3.3 Hospital admission
As a precaution, all victims of bites, scratches by snakes’ fangs or teeth, and those in whom
venom has entered the eye should be transferred as quickly as possible to the designated local
hospital, by prearranged transport, for medical assessment. It may be helpful to remove from the
cage, and take to the hospital with the victim, the label identifying the snake responsible for the
bite, so that accurate identification of the snake species and of the antivenom to be administered
is ensured.
If, as highly recommended, the appropriate antivenom is stocked by the snake farm, a supply
should accompany the victim to hospital. Hospital staff should be warned in advance by
telephone of the arrival of the casualty and informed about the species responsible and any
background medical problems and relevant medical history, such as past reactions to antivenom
or other equine sera (e.g. anti-tetanus serum), and known allergies.
An occupational hazard of snake handlers is the sensitization to venom proteins. Two out of 12
snakebites resulted in venom-anaphylaxis in a venom production plant (47). Hypersensitivity is
usually acquired by mucosal contact with aerosolized lyophilized venom. Important early
evidence of evolving sensitization is sneezing, coughing, wheezing, itching of the eyes or
weeping when entering the snake room. No one with established venom allergy should be
permitted to continue working with snakes. Venom-induced anaphylaxis should be treated with
self-injectable adrenaline (epinephrine) 0.5 ml of 0.1% solution by intramuscular injection (adult
dose) which should be stocked in the emergency drugs cupboard.
7.2.3.4 Medico-legal and health insurance aspects
The occupational exposure to venomous snakebites in commercial venom production units is the
responsibility of the employers and requires their formal attention.
7.3
26
Main recommendations
•
Well-managed snake farms are a key element in the production of venom
preparations meeting the quality requirements for the production of effective
antivenoms.
•
The quality of snake venoms used for animal immunization, as material for
preclinical assessment of neutralization efficacy, or for the development of
national or regional reference preparations is of critical importance.
•
The procedures used in snake maintenance, handling and milking, as well as in
all aspects of venom collection should be properly documented and scheduled.
•
Venoms used for antivenom preparations should be representative of the entire
snake population living in the area for which the polyspecific and/or
monospecific antivenoms are intended to be used. Because of regional and
individual variations in venom composition of snake species, the venoms used
for immunization should be collected from a large number of individuals
(generally at least 20–50) collected from various regions covering the entire
geographical distribution of the particular venomous snake species.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
•
8
8.1
Venom producers should follow the following recommendations rigorously and
should be able to demonstrate their application:
-
Taxonomic identity and geographical origin of each individual animal used
for venom production should be known and recorded.
-
Housing, feeding, and handling of snakes should meet the highest
veterinary and ethical standards, and follow documented protocols.
-
Adequate training should be provided to personnel involved in venom
production in all procedures, and implementation of health and safety
measures.
-
Formal guidelines and procedures should be established and applied in
cases where staff are bitten or have venom spat in their eyes.
-
Venom should not be milked from sick animals, which should be
quarantined.
-
Full traceability of each venom batch should be ensured.
-
Venoms should be frozen as soon as possible after collection, and at least
within 1 hour.
-
Freeze-drying or dessication of the venoms should be done under
conditions that ensure stability for long-term storage.
-
Batch-to-batch consistency of venoms of the same origin should be
confirmed.
QUALITY CONTROL OF VENOMS
Records and traceability
It is critical to identify accurately the species (and the subspecies, if any) of each individual
snake used for venom production and the taxonomic status should be validated by a competent
herpetologist. Increasingly, DNA taxonomy is replacing conventional morphological methods,
but this technique is impracticable in most venom production units which will continue to rely on
well-established physical features such as colour pattern and scale count to distinguish the
principal medically important species.
Internationally recognized scientific names should be used and the bio-geographical origin of
each snake should be specified, since differences in venom composition may occur between
different populations of the same species or subspecies (32–34, 49). Venom producers can
consult academic zoologists who have appropriate skill and experience.
Data pertaining to each numbered venom batch should include the information considered to be
key for traceability, quality and specificities of the venom (e.g. identification of all the snakes
used, the species, subspecies and biogeographical origin, feeding, health care, date of each
milking and quantity of venom produced). This information should be made available upon
request to any auditor or control authority.
In the case of long-term storage, venoms could be regularly re-dried by dessication to ensure
minimum water content, as this is critical to their long-term stability.
27
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
8.2
National reference materials
The quality of snake venoms used as a reference standard by quality control laboratories and
national regulatory authorities is crucial.
Due to the large variations in venom composition even within a single species it is recommended
that national reference venoms should be established, which cover the entire intraspecies
variability. Regional reference materials could be used when countries within the region share
similar distribution of venomous snakes.
Establishing reference venoms ensures that the antivenoms produced will be tested against the
relevant venoms in the specific countries or regions.
Venom batches may be prepared following the procedure outlined in section 7. Whatever their
origin, the snakes used for these reference standards should be accurately authenticated by a
qualified person (species, subspecies) and the place of capture recorded.
It is the responsibility of the venom producer to provide clear information on the species, the
subspecies and the geographical origin of the snakes used for the production of the venoms
supplied for antivenom production, quality control and preclinical studies. This information
should be included in the technical dossier supporting the marketing authorization of any
antivenom.
8.3
Characterization of venom batches
In addition to the certificate mentioning the scientific name of the snake species (and subspecies,
if any), the geographical origin and the number of animals used for preparing the batch, and the
date of collection of the venom, additional biochemical and biological information may be
provided for each venom batch as evidence of consistency. This information may include
analysis of:
− Biochemical characteristics of the venom:
â–¡
protein concentration;
â–¡
scans or pictures of sodium dodecyl sulfate–polyacrylamide gel electrophoresis
(SDS–PAGE) (in reducing and non-reducing conditions);
â–¡
size-exclusion chromatographic profiles (e.g. high-performance liquid
chromatography (HPLC));
− Enzymatic and toxicological activities of the venoms:
â–¡
e.g. median lethal dose, LD50.
If the venom producer is not able to perform these determinations, they can be subcontracted or,
alternatively, depending on the agreement, the antivenom manufacturer can perform relevant
assays to confirm compliance of venoms with specifications as part of the quality control system.
8.4
Main recommendations
•
28
Quality control of snake venoms is essential to provide assurance that the venoms
are representative of venomous snakes inhabiting the region for which the
antivenoms are prepared or designed.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
9
•
Traceability of each venom batch is important for rapid detection of any errors that
might occur during the preparation process.
•
For each venom batch, a certificate stating the scientific names of the snake species
(and subspecies, if any), their geographical origin and the number of animals used
in collecting the batch, the date of collection of the venom, and any other relevant
information, should be made available by the venom supplier to the antivenom
manufacturer as well as to the the regulatory authority if required.
•
Consistency, within established limits of composition and quality, of venom batches
produced over time for the same venomous species of the same origin should be
guaranteed. Specific tests should be performed and data recorded for traceability,
including: the protein concentration per g (or mg), an assessment of biochemical or
biological activity, scans or pictures from SDS–PAGE (in reducing and nonreducing conditions), and/or size-exclusion chromatographic profiles of the venom.
This information has proved useful to confirm the origin and the integrity of the
venom preparation.
OVERVIEW OF THE PRODUCTION PROCESS OF ANTIVENOMS
Antivenoms are obtained following a complex production process (Figure 1), which involves
several steps critical to quality, safety and efficacy. These steps are summarized below:
− Collection of venoms from individual venomous snakes that should be well identified
and confirmed to be in good health. They should be representative of the region(s) where
the resulting antivenom immunoglobulins are intended to be used.
− Milking of the selected snakes to prepare representative mixtures of venoms.
− Preparation of the venom(s) mixtures used for the programme of immunization of
animals (most often horses). Animals should be selected and controlled carefully, and
subjected to continuous health surveillance.
− Collection of blood or plasma from the immunized animals, once the immune response
to the immunizing venom mixture has yielded satisfactory antibody levels.
− Preparation of the pool of plasma for fractionation.
− Fractionation of the plasma to extract the antivenom immunoglobulins.
− Formulation of the bulk antivenom immunoglobulins and aseptic filling.
− Quality control tests, including potency assessment by in vivo assay.
− Labelling, packaging, boxing and release.
− Distribution within the region(s) where snakes used to prepare the venoms to immunize
the animals are prevalent.
29
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Capture of wild, well identified snakes
Snake farm
On-going veterinary surveillance
Collection of venoms (milking)
Selection of animals (eg horses)
Preparation of venom mixtures
Quarantine, vaccination &
Veterinary control
Quality control of venom mixtures
On-going veterinary surveillance
Preparation of immunizing doses of venoms
Inclusion in the herd
Immunization programme of each animal
Control of animal immune responses
Collection of blood or plasma
Storage and pooling of plasma for fractionation
Quality control of plasma for fractionation
Fractionation of plasma immunoglobulins
Formulation and filling
Quality control of antivenom immunoglobulins
Labelling, packaging and release
Figure 1
General manufacturing process of antivenoms
30
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
10 SELECTION AND VETERINARY HEALTH CARE OF ANIMALS
USED FOR PRODUCTION OF ANTIVENOMS
10.1 Quarantine period
Before an animal is introduced into the herd used for a production programme, it should be
subjected to a period of quarantine (which, in most countries, is from 6 to 12 weeks), depending
upon the source of the animal, during which an appropriate veterinarian assessment is performed
to ensure its suitability for the programme.
When an animal is imported from a country or region with different ecological characteristics, a
period of acclimatization to the local environment of about 3 months is needed. Each individual
animal should be unambiguously identified using, for example, a microchip, branding or earclipping.
In the case of horses and other equines, animals between 3 and 10 years are usually included in
an immunization programme, but in some cases older animals may also be suitable as long as
they exhibit a satisfactory immune response to the immunization programme. In the case of
sheep, animals retired from wool production have proved capable of useful antibody production
for a number of years (beyond the age of 10 years). No particular breed is preferred, but in
general large horses or sheep are preferred because they yield larger individual volumes of
blood.
10.2 Veterinary surveillance and vaccinations
The veterinary examination may include serological testing for the most prevalent infectious
diseases for that type of animal in that particular geographical location.
Depending upon the local epidemiological situation, animals should be vaccinated against
tetanus and, possibly other endemic diseases, such as rabies, equine influenza, anthrax,
brucellosis, glanders, African horse sickness and equine encephalitides. Animals should go
through a programme to eliminate gut helminths and other locally prevalent parasites.
Staff who are in regular contact with the animals should be vaccinated against tetanus and rabies.
10.3 Animal health surveillance after inclusion in the herd
After the quarantine period, if the animal is in good health according to a veterinary check-up,
and the results of relevant serological tests are negative, the animal may be incorporated into the
herd of animals used for immunization.
An individual record should be kept for each animal being used in an immunization programme
for antivenom production. In addition to surveillance by a veterinary professional, the staff in
charge of the animals should be well-trained, and the operations related to animal care and
maintenance should be clearly specified in the standard operating procedure.
During the time an animal is used for immunization aimed at antivenom production, careful
veterinary surveillance should be maintained, including continued vaccination regimes, and the
performance of regular clinical examinations, together with clinical laboratory tests such as
haemogram, clotting tests and other tests associated with the possible clinical effects of venoms
(50).
Possible anaemia, resulting from excessive volume or frequency of bleeding (when erythrocytes
are not re-infused into the animals after the whole blood bleeding session) or from the
deleterious action of venoms should also be tested for.
31
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
The immune response against venom components should, when feasible, be followed throughout
the immunization schedule, in order to detect when animals reach an acceptable antivenom titre.
However, the monitoring of the immune response can be done on a pool of sera from various
animals. This response may be followed by in vivo potency assays of neutralization of lethality
or by in vitro tests, such as enzyme immunoassays (EIAs) (provided that a correlation has been
demonstrated between these tests and the in vivo potency tests).
Whenever an animal develops any manifestation of sickness, it must be temporarily withdrawn
from immunization programmes to allow it to receive proper attention and treatment. If the
disease is controlled, the animal may return to the immunization programme after a suitable
length of time, usually 4 weeks. If an animal is receiving any sort of antibiotic or drug, it should
be withdrawn from the immunization programme for a period that would depend on the
elimination kinetics of the particular drug(s) concerned. In the case of vaccination, this
withdrawal period should not be shorter than 1 month 1 . Animals should have appropriate
physical exercise. Their feed should originate from a controlled source and should be free of
ruminant-derived material. Ideally, the diet should include both hay and grass, or alternative
plant material, and concentrated food preparations containing vitamins including folic acid, iron
and other mineral supplements. The routine quality control of the food and water is
recommended, in order to assure a consistent composition and adequate level of nutrients.
As a consequence of immunization with venoms (see section 11) a common problem in
antivenom-producing animals is the development of local ulcers or abscesses (sterile and
infected) at sites of venom injection. This is a particular problem when necrotic venoms and
complete Freund’s adjuvant are used. All injections should be given under aseptic conditions.
There should be a limit to the total volume and dose of venom injected at a single site. Infected
or ulcerated areas should not be used again until they have fully healed. In the event of the death
of an animal being used for antivenom production, a careful analysis of the causes of death
should be performed, including, when necessary, the performance of a necropsy.
Some animals show declining titres of specific venom antibodies over time, despite rest or
increasing doses of immunizing venoms. Such animals should be retired from the immunization
programme. In agreement with GMP principles and to avoid impact on the composition and
consistency of the antivenom produced, it is, in principle, not considered good practice to move
animals from a given venom immunization programme to another one, unless the animal has
been used in the preparation of a monospecific antivenom that is included into a polyspecific
preparation, or if it was used for the production of other animal-derived antisera (e.g. anti-rabies,
anti-tetanus, or anti-botulism).
10.4 Main recommendations
•
Animals intended for antivenom production programmes should be identified to
ensure full traceability and health surveillance.
•
Animals should go through a quarantine period of 6–12 weeks during which they
are submitted to veterinary scrutiny and are vaccinated against and treated for
parasites.
•
Following the quarantine period, they are introduced into the immunization
programme. Animals should be appropriately housed, fed, and managed according
to the highest veterinary and ethical standards.
1
In some areas, legislation stipulates that animals used for production of plasma cannot be treated with
penicillin or streptomycin.
32
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
•
During immunization, the clinical status of each animal must be followed by a
veterinarian through clinical and laboratory assessments. If an animal develops
signs of disease, it should be temporarily separated from the immunization
programme to receive appropriate treatment. Particular care must be paid to the
local lesions that develop at the site of venom injections.
•
The immune response to venoms of each animal should, when possible, be
monitored during the immunization schedule; alternatively, the antivenom titres
can be monitored indirectly by testing the plasma pool.
•
An animal receiving an antibiotic or drug should be withdrawn from the
immunization programme for a period depending on the elimination kinetics of
each drug. In the case of vaccination, this withdrawal period should not be shorter
than 1 month.
11 IMMUNIZATION REGIMENS AND USE OF ADJUVANT
One of the most crucial steps in antivenom production involves the immunization of animals
with venom(s) to produce a long-lasting and high titre antibody response against the lethal and
other deleterious components in the immunogenic toxins. To achieve this goal, the following
considerations are important:
− Venom(s) used should be prepared as described in section 7, and should be in an optimal
condition for inducing specific and neutralizing antibodies.
− Immunogen and the immunization regimens used should not seriously affect the health
of the animal.
− Preparation of immunogens and the immunization protocol should be technically simple
and economical and use a minimal amount of venom. The procedures followed must be
included in a protocol and their performance must be documented.
The antivenom manufacturer is responsible for defining the appropriate immunization
programme (choice of doses, selection of adjuvants, sites of immunization, and bleeding
schedule) able to generate the best immune response and plasma production, while also ensuring
optimal animal care. GMP principles should be applied in the preparation of the immunizing
doses as well as in the immunization process.
11.1 Animals used in antivenom production
Numerous animal species have been used on various scales in antivenom production (horse,
sheep, donkey, goat and rabbit) or for experimental purposes (camel, llama, dog and hen) (51,
52). However, the production of large volumes of antivenom from large animals such as equines
is an advantage compared to the smaller species. The selection of the animal species should be
based on several considerations, such as locally prevalent diseases, availability in the region,
adaptation to the local environment, and cost of maintenance. The information in these
Guidelines refers mostly to horse-derived immunoglobulins.
The horse is the animal of choice for commercial antivenom production. Horses are docile,
thrive in most climates and yield a large volume of plasma. Antivenoms made from horse plasma
have proven over time to have a satisfactory safety and efficacy profile (53). Sheep have also
been used as an alternative source for antivenom production because they are cheaper, easier to
raise, can better tolerate oil-based adjuvant than horses, and their antibodies may be useful in
patients who are hypersensitive to equine proteins. However, increasing concern about prion
33
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
diseases may limit the use of the sheep as an animal for commercial antivenom production.
Larger animals are preferable to smaller ones because of their greater blood volume, but breed
and age are less important. Any animals used should be under veterinary supervision (see section
10). When sheep or goats are to be used, manufacturers should comply with regulations to
minimize the risk of transmissible spongiform encephalopathies to humans, such as the WHO
Guidelines on tissue infectivity distribution in transmissible spongiform encephalopathies (54).
11.2 Venoms used for immunization
Venoms used as immunogens in antivenom production are chosen based on criteria discussed in
section 6. Priority should be given to venoms from snakes responsible for frequent envenomings.
The quality, quantity, and biological variation of venoms are important considerations (see
sections 7 and 8).
11.3 Preparation of venom doses
Venom doses used for the immunization of animals should be prepared carefully in a clean
environment, with an established, scheduled and documented cleaning regime. All venom
manipulations should be performed using aseptic techniques under a hood; for highly toxic
venoms, a cytotoxic cabinet may be used. Batch process records should be completed for each
dose preparation session. The venom batches used and the animals to be immunized should be
recorded and the containers in which the venom is dissolved should be appropriately identified.
Ideally, the calculations and operations related to the dose of venom to be used, as well as
dilutions, require verification by a second person to ensure accuracy and to prevent errors that
may lead to animals receiving overdoses.
Venoms, when freeze-dried, are highly hygroscopic and allergenic, thus care should be taken
when manipulating them. When taken out of the refrigerator or freezer, the venom should be
allowed to warm up to room temperature before the bottle is opened, otherwise condensation
may occur causing inaccuracy in weighing and, more seriously, proteolytic degradation of the
venom proteins by venom enzymes. Venom should be dissolved in distilled water or buffer, but
care should be taken not to shake the solution too vigorously since excessive foaming may cause
protein denaturation.
The solvents used to dissolve venoms should be sterile and within established expiry periods. A
stock solution of each venom should be prepared separately, rather than being mixed with other
venoms. This is to allow flexibility of dosage and to avoid proteolytic degradation by one venom
component of other venom proteins. Venom solutions can be sterile-filtered where this is known
not to affect the potency of the preparation, aliquoted, labelled and stored appropriately (e.g.
refrigerated, frozen at –15 to –20 °C, or deep frozen at –70 °C) for a short time (less than 1
month). However, it is recommended that venoms used for immunization be freshly prepared at
the time of use.
All the equipment used for venom storage (freezers and refrigerators) and preparation (e.g.
balances) should be calibrated and validated for their intended purpose. Balances should be
calibrated at least annually and calibration should be checked daily. Where possible, laboratory
items used in venom preparation, i.e. pipettes, syringes and other such items should be presterilized, single-use, disposable items. The siliconization of venom solution containers may be
considered to avoid the adherence of venom components to the surfaces of containers. Transport
of venom solutions to the facilities where animals are going to be injected should be done in a
safe manner.
34
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Care should be taken to avoid accidents that may result in envenoming of the persons preparing
the venom solutions. Protective equipment (e.g. eyewear, gloves and gowns) should be worn by
personnel preparing venom solutions. Procedures for cleaning up broken glass or plastic
containers that have held venom should be established and the personnel should be trained to
follow them.
11.4 Detoxification of venom
Some snake venoms can cause local and/or systemic toxicity when injected into naive horses at
the beginning of an immunization course. Various physical or chemical means have been
adopted to decrease venom toxicity, for example, treatment with aldehydes (formaldehyde or
glutaraldehyde), hypochlorite, ultraviolet or gamma radiation, and heat, among others. However,
in most cases, not only the toxic sites, but also the antigenic sites of the toxins are destroyed after
these treatments (55). For example, when glutaraldehyde is used, the protein polymerization is
often extensive and is difficult to control and reproduce. Thus, although the detoxified toxin
(toxoid or venoid) induces vigorous antibody response, the antibodies usually fail to neutralize
the native toxin. In fact, no detoxification is usually necessary if inoculation is made with a small
dose of venom well-emulsified in an adjuvant such as Freund’s complete or incomplete
adjuvants.
11.5 Immunological adjuvants
Various types of immunological adjuvants have been tested, for example, Freund’s complete and
incomplete adjuvants, aluminium salts (hydroxide and phosphate), bentonite and liposomes (56).
The choice of adjuvant is determined by its effectiveness, side-effects, ease of preparation,
especially on a large scale, and cost. It may vary depending upon the type of venoms and
following manufacturers’ experience. Freund's incomplete adjuvant (FIA) contains mineral oil
and an emulsifier. Freund’s complete adjuvant (FCA), which contains mineral oil, an emulsifier
and inactivated Mycobacterium tuberculosis, has been shown in experimental animals to be one
of the most potent adjuvants known. However, horses are quite sensitive to FCA which tends to
cause granuloma formation. For this reason, some producers prefer to use other adjuvants.
It has been noted that the granuloma caused by FCA is due to injection of a large volume (5–10
ml) of the emulsified immunogen at 1 or 2 sites. The large granuloma formed usually ruptures,
resulting in a large infected wound. If the emulsified immunogen is injected subcutaneously in
small volumes (50–200 µl/site) at multiple sites of injection, granuloma formation may be
avoided.
11.6 Preparation of immunogen in adjuvants
To minimize infection at the immunization sites, all manipulations should be carried out under
aseptic conditions. Venom solutions are prepared in distilled water or phosphate-buffered saline
solution (PBS) and filtered through a 0.22-µm membrane. The venom solution is then mixed
and/or emulsified with adjuvant, according to the instructions of the supplier. An example for the
preparation of venom immunogen in FCA, FIA and aluminium salts is described in Box 1.
35
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Box 1
Example of preparation of venom immunogen in FCA, FIA and aluminium salts
Since FCA can cause severe irritation, precautions should be taken to avoid contact with the
eyes, and protective eyewear and gloves are recommended. The vial containing FCA is
shaken to disperse the insoluble Mycobacterium tuberculosis. The venom solution is mixed in
a stainless steel container with an equal volume of FCA at 4 ºC. The emulsification is
achieved by vigorous blending in a high-speed blender at a speed of approximately 3000 rpm
for 15 minutes. The container is put in ice water to dissipate the heat generated. The resultant
emulsion should be quite thick and remains stable when dropped on the surface of cold water.
The highly viscous emulsion is then transferred into a sterile 50-ml glass syringe with the
plunger removed. The plunger is then put into the syringe to expel any air pocket inside. By
means of a three-way stopcock, the emulsion is then transferred from the 50-ml syringe into a
1-ml or 2-ml glass syringe. With the use of an 18G–20G needle, the water-in-oil emulsion is
ready for subcutaneous injection.
Immunogen in FIA is prepared by a process similar to that described above except that FIA is
used in place of FCA. Both the FCA and FIA emulsified immunogens may, if necessary, be
stored at 4 ºC, preferably for a maximum of 2 weeks but re-emulsification is needed before
their injection.
When the immunogen is prepared in Al(OH)3 (aluminium hydroxide) or Al(PO)4 (aluminium
phosphate), a sterile venom solution and a suspension of aluminium salts are mixed in a ratio
of 1:3 (v/v) and homogenized. When using other adjuvants, the preparation of the solution or
emulsion should follow the manufacturer’s instructions for that type of adjuvant.
11.7 Immunization of animals
The areas to be immunized should be thoroughly scrubbed with a disinfectant, shaved and
rubbed with 70% ethanol before venom immunogen injection.
In general, the sites of immunization (Figure 2) should be in areas close to major lymph nodes,
preferably on the animal’s neck and back, while the route of injection should be subcutaneous so
as to recruit a large number of antigen presenting cells and consequently resulting in high
antibody response. Some procedures call for a small volume of injection at each site (50–200 µl)
so that the total surface area of the immunogen droplets is maximized, enhancing the interaction
with the antigen presenting cells and the immune response (57, 58). An example of
immunization of a horse using venom emulsified in FCA is described in Box 2.
Other immunization protocols, using larger amounts of venoms devoid of local tissue-damaging
activity (such as those of some elapids) and/or adjuvants other than FCA may be used with
satisfactory results, as long as the schedule does not compromise the health of the animals. In
situations where the main toxins of a given venom have a low molecular mass and would not
induce a sufficient immune response if injected together with the other venom components,
isolating such toxins using mild chromatographic procedures can be beneficial. Such isolated
fractions can then be used for immunization.
36
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Figure 2
Recommended areas of immunization in horses
37
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Box 2
Example of immunization of horses using FCA, FIA and aluminium salts
The primary immunization could be made with venom(s) mixed with (FCA) as described in
Box 1. The initial dose of each venom could be as low as 1–4 mg/horse with a total combined
volume of injection of about 2 ml. The immunogen is filled in a 1-ml glass syringe with an
18G needle as described above. Subcutaneous injections of 100–200 µl of immunogen are
made at each site, up to as many as 8–12 sites, although some producers may use only 3–4
injection sites. The neck of the horse, supplied with extensive lymphatic vessels and large
lymph nodes, is a preferred area for immunization. If inoculation is made on the lateral sides
of the neck, the animal tends to rub itself causing skin blisters. Thus, injections should be
made to the upper (dorsal) part of the neck, close to the mane. About 4–6 injections can be
made at each side of the neck. If injection in the rump is possible, 1–2 injections can be made
in the area between the outer hip bone and the top of the thigh bone. The scratching of
injected sites by animals can be partially alleviated by massaging the injection site after
venom injection to disperse the dose material.
Immunization using Freund’s complete adjuvant is usually made only once; repeated use of
this adjuvant may in most cases cause serious reactions which can affect the horse’s health.
After 2 weeks, the horses should receive a booster injection with the same venom(s) well
emulsified in Freund’s incomplete adjuvant. Similar volume and areas of injection to those
described above can be made. Subsequent booster immunizations at 2-week intervals can be
made with higher doses (5–10 mg) of venom(s) in saline or mixed with aluminium salts or
any other adjuvant selected. In this case, subcutaneous injections of 1 ml of immunogen at
each site in a total of 4 sites are recommended.
Blood (10–20 ml) should be drawn before each immunization. Serum or plasma is prepared
and EIA (enzyme immunoassay) titres and/or lethality potency are determined. When the EIA
titres reach a plateau, usually about 8–10 weeks after the primary immunization, an in vivo
potency assay may be performed to confirm that the horse could be bled. After bleeding for
antivenom production, the horses are allowed 3–8 weeks rest, depending on their physical
condition. After the rest period, a new round of immunization can be made as described
above, but without the use of Freund’s complete adjuvant.
11.8 Traceability of the immunization process
The traceability of the immunization process is critical for the quality control of the antivenoms
produced and the steps to ensure it should be performed very accurately. Each immunized animal
should be identified by its code number (see section 10). The details of each immunization step
should be recorded precisely. The details to be recorded include:
— date of immunization;
— batch(es) of venom(s) used with its (their) reference number(s) (see section 8);
— venom dose(s);
— adjuvant and/or salt used;
— names of the veterinary and supporting staff in charge of the immunization;
— eventual reaction and/or sickness.
38
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
The antivenom titre of the immunized animals should be followed throughout the immunization
procedure either in vitro, using EIA, during the immunization phase, or in vivo, by neutralization
potency assays of lethality when the immunization plateau is reached or before each blood
collection.
Each plasma batch should be assigned a unique reference number (e.g. a bar code), which should
allow complete traceability to the donor animal. Information (such as the date of collection, the
unique identification number of the immunized donor animal, and the reference number of the
venom(s) used for immunization) should be recorded to allow traceability to all venoms.
Computer-based databases are very useful for properly recording these data, which are crucial
for the traceability of the antivenoms produced. Standard procedures should be used to protect
the integrity of data stored on a computer, including regular, frequent backup, protection from
unauthorized access, and storing backup copies securely off-site.
11.9 Main recommendations
•
Venom solutions should be prepared in such a way as to minimize proteolytic
digestion and denaturation of the venom proteins. Venom solution should be
prepared under aseptic conditions to avoid infection at the injection sites.
•
The type of adjuvant used is selected on the basis of its effectiveness, side-effects,
ease of preparation and cost.
•
Primary immunization should be made by subcutaneous injections of small volumes
at multiple sites close to the animal’s lymphatic system to favour the recruitment of
antigen presenting cells and involving anatomically different groups of lymph nodes
for antibody production.
•
Subsequent booster injections can be made using venom immunogen doses, at
volumes and intervals depending on the type of adjuvant used, until the antivenom
titre reaches a plateau or a pre-established minimum accepted titre.
•
After collection of blood for antivenom production, animals should have a resting
period of 3–8 weeks. After this, a new round of immunization can be performed as
above without the use of Freund’s complete adjuvant.
•
All steps in the immunization of the immunized donor animal, as well as the
collection of blood or plasma should be traceable.
12 COLLECTION AND CONTROL OF ANIMAL PLASMA FOR
FRACTIONATION
Historically, serum separated from the blood of hyperimmunized horses was the basis of
“antivenin serum-therapy”, but today plasma is used, almost exclusively, as the starting material
and undergoes a fractionation process for the separation of purified antivenoms. Thus
“antivenom immunoglobulins” is the preferred term, rather than “anti-snakebite serum” or
“antiserum” which are imprecise and confusing terms that refer to a crude therapeutic
preparation.
Plasma as a starting material is preferred to serum largely because erythrocytes can be returned
to the animal, thus preventing anaemia and hypovolaemia in the donor animal and allowing more
frequent bleeding. Some laboratories have found that using plasma enables higher recovery of
antibodies per donation and it is less contaminated with haemoglobin. Separation of plasma from
anticoagulated blood is much more rapid than separation of serum from clotted blood. Plasma for
39
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
fractionation can be obtained either from the collection of whole blood or by the apheresis
procedure.
12.1 Health control of the animal prior to and during bleeding sessions
When an immunized animal has developed an antivenom antibody titre that meets the necessary
specifications, it can be bled. Before bleeding is performed, the animals should be evaluated by a
veterinarian or other qualified person and declared healthy. Animals showing evidence of
clinical deterioration, such as weight loss, drop in haemoglobin or serum protein concentration
below a critical predefined value, or evidence of infections, should not be bled.
12.2 Premises for blood or plasma collection
The bleeding of animals should be performed in designated rooms or areas dedicated to this
activity and equipped with appropriate restraining devices. Some producers may design the
bleeding rooms so that they can be closed, if needed, during the bleeding sessions, but this is not
general practice. The rooms or areas should be thoroughly washed and cleaned before and after
each bleeding session and their design should facilitate such cleaning procedures, which should
be clearly established. The room or area should be inspected before the confinement of the
animal. Animals need to be made as safe and comfortable as possible, in a quiet environment,
during bleeding to minimize the chance of injury to the animal or its handlers. Individual animals
should be confined in circumstances that reduce the potential for stress as much as possible. It is
recommended that these rooms allow the simultaneous bleeding of various horses to reduce the
time required for this operation as well as the stress.
12.3 Blood or plasma collection session
Animals are bled by venipuncture from the external jugular vein. The area surrounding the
venipuncture site should be shaved before bleeding and thoroughly cleaned and disinfected,
using a disinfectant that has not reached the end of its recommended shelf-life, and, depending
on the type of disinfectant, it should be allowed to dry. The disinfected area should not be
touched or palpated before the needle has been inserted.
Before venipuncture all containers and tubing should be inspected for defects (for example,
abnormal moisture or discoloration as these may suggest a defect). There should be means to
determine the volume of blood or plasma collected (such as a weighing machine).
The clinical condition of the animals being bled should be closely monitored at the time of
bleeding and during the days that follow, and bleeding should be suspended in the event of any
adverse effect on the animal. If an animal shows signs of distress during the operation, the
collection procedure should be terminated. In addition, animals should be kept under observation
for at least 1 hour after the bleeding to allow any evidence of physical alterations to be detected.
12.4 Labelling and identification
The identity of the animal should be recorded immediately before venipuncture. Labels on all
bottles or bags of blood or plasma should be marked with the animal’s unique identification
number. The label should contain the following information: specificity of antivenom, plasma
unit number and date of collection.
40
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
12.4.1 Collection and storage of whole blood
12.4.1.1 Collection
The volume of blood to be obtained depends on the species and size of the immunized animal. It
is recommended that around 13–15 ml of blood per kilogram body weight are collected in one
bleeding session. For sheep, 0.5 l is a typical yield, whereas in the case of horses, the volume of
blood may range between 3 and 6 l, depending on the size of the animal.
Blood is collected, ideally, in disposable plastic bags containing sterile citrate anticoagulant.
Usually, the volume ratio of anticoagulant to blood is 1:9. Use of double plastic bags containing
anticoagulant is recommended to avoid bacterial contamination and for ease of use. When plastic
bags are not available, disposable polypropylene plastic bottles, or sterilized glass bottles
containing anticoagulant may be considered.
While the bleeding is taking place, a constant flow of blood should be ensured. Blood should be
gently and continuously mixed with the anticoagulant solution to ensure a homogeneous
distribution of the anticoagulant, to avoid the risks of activation of the coagulation cascade and,
therefore, avoid the formation of clots. The duration of a bleeding session is usually between 30
and 45 minutes depending upon the weight of the animal and the total volume collected. Care
should be taken to avoid contamination of the blood by exposing the needle to contaminated
surfaces.
12.4.1.2 Storage
The bags or bottles in which the whole blood has been collected should be appropriately cleaned
and sanitized on their external surfaces. They should be put into a refrigerated room (2–8 °C) for
the plasma and blood cells separation procedure. They should be stored for not more than 24
hours before the reinfusion of the red cells.
Alternatively, aseptically-collected blood can be stored for a maximum of 7 hours at 20–25 °C to
allow for sedimentation. Under such circumstances, great care should be taken to avoid bacterial
contamination.
12.4.1.3 Separation of plasma from whole blood
Hyperimmune plasma should be separated from blood cells under aseptic conditions and should
be transferred into sterile containers (plastic bags, bottles, or stainless steel containers). A
designated room, designed to allow proper cleaning and sanitization, should be used for
separation. When bottles are used, separation of plasma from blood cells should be performed in
a laminar flow cabinet located in a room separated from the plasma fractionation area.
12.4.1.4 Reinfusion of the erythrocytes
Reinfusion of the erythrocytes after whole blood collection is recommended.
Blood cells, most specifically erythrocytes (red blood cells), should be separated from plasma by
validated centrifugation or sedimentation procedures. Erythrocyte reinfusion should take place
within 24 hours after blood collection, and after being suspended in sterile saline solution at 32–
37 °C prior to infusion. This procedure in which whole blood is collected and erythrocytes are
reinfused to the animal is commonly referred to as “manual apheresis”.
41
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
12.4.2 Plasma collection by automatic apheresis and storage
12.4.2.1 Plasma collection
In some laboratories, plasmapheresis machines are used to perform automatic plasma collection.
This has proved a useful investment in some facilities; it ensures that the animal does not
become hypovolaemic and it reduces the risks of handling errors, in particular during re-infusion
of the erythrocytes to the donor. Plasma from automatic apheresis tends to be less contaminated
by blood cells (red blood cells, leukocytes and platelets) and in the experience of some
laboratories is easier to fractionate, as the filtration steps, in particular, are more readily
performed, resulting in higher yields.
In such procedures, whole blood is collected from the animal, mixed with anticoagulant, and
passed through an automated cell separator. The plasma is separated from the cellular
components of the blood, which are returned to the animal in a series of collection/separation
and return cycles. The plasma is separated from the red blood cells by centrifugation or filtration,
or a combination of the two. The operational parameters of the plasmapheresis equipment are
provided by the manufacturers of the equipment. In general, the anticoagulant is delivered at a
rate yielding a specified ratio of anticoagulant to blood. The anticoagulant solutions used include
AB16 (35.6 g sodium citrate, 12.6 g citric acid monohydrate, 51.0 g glucose monohydrate per
1 litre using water for injection) and anticoagulant citrate dextrose formula A (ACDA) (22.0 g
sodium citrate, 8.0 g citric acid, 24.5 g dextrose monohydrate, per 1 litre using water for
injection). The number of collection/separation and return cycles for each donor animal depends
on the total volume of plasma that is to be harvested. For horses, the average volume of plasma
collected may be about 6 litres per session. The number of cycles ranges from 10 to 20
depending upon the haematocrit of the horses. The collection process lasts for 1–4 hours. The
apheresis equipment and apheresis procedures should be validated, maintained and serviced.
Machine plasmapheresis can take several hours and animals can be fed during the operation.
12.4.2.2 Plasma storage
Apheresis plasma: bags or bottles should be stored in a refrigerated room (2–8 °C) in the dark
until the fractionation process starts. This storage room should be designed to allow proper
cleaning and sanitization.
12.5 Pooling
Plasma from individual animals should be pooled into sterile and sanitized containers before
fractionation. For traceability purposes each plasma pool should be identified with a unique
number. The number of plasma units collected from individual animals and used in the pool
should be recorded.
Such pooling should be performed in an environment suitable to prevent microbial
contamination, like classified areas (class D (59)) and pools should be adequately identified. The
room should be designed to allow for appropriate cleaning and sanitization of all surfaces.
Individual or pooled plasma should be stored at 2–8 °C in a room dedicated for this purpose. To
ensure the prevention of microbial contamination of plasma, preservatives (phenol or cresols)1
can be added at a dose of less than 3 g/l at this stage and kept during storage of plasma. Care
should be taken to dilute the phenol or cresols with water or saline solution before they are added
to plasma, to avoid denaturation of plasma proteins. The transportation of containers or bottles
1
42
In these guidelines, cresol isomers are referred to as cresols.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
containing pooled plasma within the production facility or between facilities should be
performed in such a way that contamination is avoided and the cold chain is maintained.
To avoid the risk of contamination, it is recommended that individual or pooled plasma is not
stored for too long before fractionation, i.e. the plasma should be fractionated as soon as possible
after pooling. In the event that plasma is stored for prolonged periods of time (for instance
6 months), the storage time and conditions should be validated to ensure that there is no
detrimental impact on the quality of the plasma material, on the fractionation process, or on the
quality, efficacy and stability of the antivenoms.
It is also the experience of some manufacturers that plasma can be stored frozen at −20 °C,
particularly if no preservative is added.
12.6 Control of plasma prior to fractionation
Before fractionation, pools of plasma should be checked for macroscopically evident
precipitates, gross haemolysis and bacterial contamination (bioburden assay). The neutralizing
potency of the starting plasma should be ensured so that the resulting antivenoms will be within
potency specifications. Additional checks may include, when relevant, a test for pyrogenic
substances and total protein content.
Plasma pools should be discarded if the bioburden exceeds a defined limit stated in the
marketing dossier or if the neutralizing potency is below a minimum limit established by the
producer. Cloudy plasma, below this defined bioburden limit, may still be used for fractionation
provided the fractionation process and product quality has been proven not to be impaired.
Grossly haemolysed plasma should not be used for fractionation.
12.7 Main recommendations
•
When animals have developed an adequate immune response against venoms, and if
they are in good health, they can be bled for antivenom production. Bleeding should
be performed in enclosed rooms which should be kept scrupulously clean.
Traceability of the donations should be ensured.
•
Plasma is preferred to serum as a source material. Animals should be bled from the
external jugular vein. Plasma can be obtained either from whole blood or by
automated plasmapheresis and using approved anticoagulants. Blood or plasma
should ideally be collected into closed plastic bags. When this is not possible, glass or
plastic bottles can be used, if they can be readily cleaned and sterilized.
•
Plasmapheresis is recommended using either automatic or manual procedures.
When manual apheresis is used, blood cells should be sedimented, separated from
the plasma, resuspended in saline solution and returned to the animals within
24 hours. Plasma separation should be performed in a designated room with a
controlled environment.
•
Plasma containers should be thoroughly cleaned on their external surfaces,
adequately identified and stored in refrigerated rooms for further fractionation.
•
Plasma should be checked prior to fractionation to establish compliance with
relevant acceptance criteria for fractionation, in particular the neutralizing potency.
•
Special attention should be paid to ensuring traceability between individual animal
donors and the plasma pool.
43
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
•
A certificate from a veterinarian or other qualified person should be issued stating
that the donor animals were checked periodically to ensure that they were in good
health at the time of plasma collection and during the follow-up observation period.
13 PURIFICATION OF IMMUNOGLOBULINS AND IMMUNOGLOBULIN
FRAGMENTS IN THE PRODUCTION OF ANTIVENOMS
13.1 Good manufacturing practices
The purification of immunoglobulins and immunoglobulin fragments for the production of
antivenoms should aim at obtaining products of consistent quality, safety and efficacy. The
fractionation processes used should adhere to the GMP principles developed for medicinal
products. All operations should therefore be carried out in accordance with an appropriate
quality assurance system and GMP. This covers all stages leading to the finished antivenoms,
including the production of water, the production of plasma (animal selection and health control,
production of venoms and immunization protocols, containers used for blood and plasma
collection, anticoagulant solutions and quality control methods) and the purification, storage,
transport, processing, quality control and delivery of the finished product. Of particular relevance
is the control of microbiological risks, contamination with particulates and pyrogens, and the
existence of a documentation system that ensures the traceability of all production steps. To
establish satisfactory traceability of the antivenom produced, all the steps of the purification
procedure used for the preparation of the antivenom batch should be recorded carefully in preestablished and approved batch record documents, and sampling should be done at established
critical steps for in-process quality control tests.
WHO Guidelines on good manufacturing practices for medicinal products are available (59) and
the main principles of GMP for the manufacture of blood plasma products of human origin have
also been published (60, 61). These Guidelines can serve as a general guide for manufacturing
practices in the production of antivenoms. A useful reference in the field of antivenoms is also
the Note for guidance on production and quality control of animal immunoglobulins and
immunosera for human use (CPMP/BWP/3354/99) (62).
13.2 Purification of the active substance
Antivenoms are prepared from the starting plasma pool using diverse methods to obtain one of
the following active substances:
— intact IgG molecules;
— F(ab')2 fragments; or
— Fab fragments.
In general, fractionation procedures should not impair the neutralizing activity of antibodies; it
should yield a product of acceptable physicochemical characteristics and purity with a low
content of protein aggregates, which is non-pyrogenic and which should provide good recovery
of antibody activity.
The characteristics of a batch of plasma to be fractionated should be clearly established, and the
methods used to purify the active substance and the in-process controls should be described in
detail in standard operating procedures. In the following sections, examples of basic protocols
used for the production of IgG, F(ab')2 and Fab antivenoms are described. Some additional
methodologies introduced to further purify the active substance of antivenoms are also discussed.
Variations in these manufacturing procedures have often been developed by individual
44
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
fractionators and should be considered as acceptable when shown to yield consistently safe and
effective preparations of antivenoms.
13.2.1 Purification of intact IgG antivenoms
13.2.1.1 Ammonium sulfate precipitation
In the past, most laboratories have used fractionation protocols based on salting-out procedures
employing ammonium sulfate or sodium sulfate (63). Two precipitation steps are included using
two different salt concentrations in addition to the elimination of “euglobulins” by precipitation
in a diluted acidic solution.
Such fractionation protocols generally lead to a recovery of antibodies of between 40 and 50%
and to the formation of protein aggregates. The final product of this procedure used to contain a
relatively high proportion of contaminating proteins, such as albumin (64). This compromised
the safety of the product, since a high incidence of early adverse reactions has been described in
response to such intact IgG antivenoms (65).
13.2.1.2 Caprylic acid precipitation
The use of caprylic acid (octanoic acid) as an agent for precipitating proteins from animal plasma
has been described in the literature (66). Several procedures for the purification of whole IgG
antivenoms with a good physicochemical profile and purity using caprylic acid precipitation of
non-immunoglobulin proteins have been developed (64, 67, 68) and are now used for the
production of licensed antivenoms.
Figure 3 illustrates a particular process in which caprylic acid is added slowly to undiluted
plasma, with constant stirring, to reach a concentration of 5% (v/v) and pH 5.5. The mixture is
stirred at 22–25 °C for a minimum of 1 hour. The precipitated proteins are removed by filtration
or centrifugation and discarded. The filtrate or the supernatant containing the immunoglobulins
is then submitted to tangential flow filtration to remove residual caprylic acid and lowmolecular-mass proteins, depending on the molecular cut-off of the ultrafiltration membranes,
and to concentrate the proteins. The immunoglobulin solution is then formulated by adding
sodium chloride solution (NaCl), an antimicrobial agent and any other excipient(s) needed, such
as stabilizers. The pH is then adjusted to a neutral value and finally subjected to sterile filtration
through a filter of pore size 0.22 µm, and dispensed into final containers (vials or ampoules).
Variations of this procedure have been introduced by various manufacturers, and include dilution
of plasma, changes in caprylic acid concentration, pH, and temperature among others.
Caprylic acid fractionation allows the production of antivenoms of relatively high purity and
with a low protein aggregate content, because the immunoglobulins are not precipitated. The
yield may reach up to 60–75% of the activity in the starting plasma, depending upon the details
of the procedure and/or the equipment used. The efficacy and safety profiles of caprylic acidfractionated antivenom immunoglobulins have been demonstrated in clinical trials (65, 69).
45
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Figure 3
Example of a fractionation process in which intact IgG is prepared by caprylic acid
precipitation of non-immunoglobulin proteins
46
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
13.2.2 Purification of F(ab')2 antivenoms
Many manufacturers follow the classical protocol for F(ab')2 antivenom production developed by
Pope (6, 7), with a number of recent modifications (9, 10, 70).
The method of pepsin digestion (see Figure 4) involves the digestion of horse plasma proteins by
pepsin, leading to the degradation of many non-IgG proteins, and to the cleavage of IgG into
bivalent F(ab')2 fragments by removal and digestion of the Fc fragment into small peptides. A
heating step and the purification of F(ab')2 fragments by salting-out using ammonium sulfate are
also key elements of this methodology. Some procedures involve performing the pepsin
digestion step on a pre-purified IgG fraction that is obtained by treatment of plasma with
ammonium sulfate to obtain an IgG-enriched precipitate, whereas albumin is not precipitated.
Pepsin digestion is accomplished at a pH of 3.0–3.5. A typical protocol is based on incubation at
pH 3.3 for 1 hour, at 30–37 °C in a jacketed tank, with a pepsin concentration of 1 g/l. Other
procedures can be used which give similar results. Each manufacturer should adjust the pepsin
concentration to achieve the required enzymatic activity.
13.2.2.1 Downstream processing using ammonium sulfate
After pepsin digestion, the pH is adjusted to 4.5–5.0, by adding NaOH or a weak alkaline buffer;
then ammonium sulfate is added with stirring to a final concentration usually close to 12% (w:v).
The precipitate is eliminated by filtration or centrifugation, and the filtrate, or supernatant, is
heat-treated (usually at 56 °C for 1 hour; this is known as “thermocoagulation”). After
thermocoagulation, the preparation is cooled down to less than 30 °C, e.g. by passing cold water
through a jacketed vessel. The resulting fraction is filtered or centrifuged to remove the
precipitate. The pH is then adjusted to 7.0–7.2 with NaOH, and a solution of ammonium sulfate
is added with stirring to a final concentration high enough to precipitate the F(ab')2 fragments
(usually 23% (w:v) or higher). After an additional filtration step, or following centrifugation, the
F(ab')2 precipitate is dissolved, and then desalted (to remove the ammonium sulfate) and
concentrated preferentially by tangential flow diafiltration. Care should be taken to avoid
aggregate formation by ensuring gentle mixing and rapid dissolving of the precipitate.
Alternatively, the 23% (w:v) step is bypassed by some manufacturers and, directly after the
heating step, the filtrate obtained is subjected to ultrafiltration. Additional precipitation could also
be applied on the starting material at a low ionic strength and acid pH to remove "euglobulins"
(10).
The F(ab')2 solution is then formulated by adding NaCl, an antimicrobial agent, and any other
excipient needed for formulation, such as protein stabilizers, and the pH is adjusted, generally to
a neutral value. Finally, the preparation is sterilized by filtration through 0.22-µm filters, and
dispensed into final containers (vials or ampoules). Such a process, or similar ones developed by
other manufacturers, using pepsin digestion, ammonium sulfate precipitation and tangential
diafiltration is the most often used for the manufacture of F(ab')2 fragments. The yield of this
fractionation protocol usually ranges between 30% and 40%.
13.2.2.2 Downstream processing using caprylic acid
Purification of F(ab')2 has also been shown, on an experimental scale, to be achievable by
caprylic acid precipitation of non-F(ab')2 proteins after pepsin digestion, with an improved yield
(∼60%) (71). However, the yield obtained on a large scale has not been reported. Figure 5 shows
a fractionation scheme of F(ab')2 using caprylic acid. F(ab')2 is not precipitated, therefore
reducing the formation of aggregates. Some manufacturers have introduced additional processing
steps such as ion-exchange chromatography or ultrafiltration to eliminate low-molecular-mass
contaminants.
47
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Figure 4
Example of a fractionation process in which F(ab')2 fragments are prepared by pepsin
digestion and ammonium sulfate precipitation
48
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Figure 5
Example of a fractionation process in which F(ab')2 fragments are prepared by pepsin
digestion and caprylic acid precipitation
49
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
13.2.3 Purification of Fab antivenoms
Production of monovalent Fab fragments is performed by some manufacturers (72), currently
using hyperimmunized sheep plasma. Papain is used to carry out the enzymatic digestion, and
the process of preparation of the fragment may use ammonium sulfate, sodium sulfate or caprylic
acid.
Figure 6 shows a process in which immunoglobulins are precipitated from plasma by adding
ammonium sulfate or sodium sulfate to a concentration of 23%. After filtration the filtrate is
discarded and the immunoglobulin precipitate is dissolved in a sodium chloride solution at
pH 7.4. Papain is added and digestion performed at 37 °C for 18–20 hours in a jacketed tank.
Reaction is stopped by adding iodoacetamide. The product is then applied to a diafiltration
system to remove iodoacetamide, salts and low-molecular-mass peptides and equilibrated with a
buffered isotonic NaCl solution. The preparation is then chromatographed on an anionexchanger (usually in quaternary aminoethyl (QAE)-based or diethylaminoethyl (DEAE)-based
media). Fc fragments and other impurities are bound on the column, whereas Fab fragments pass
through. After an additional diafiltration/dialysis step, the product is formulated by adding NaCl,
antimicrobial agents, when used and any other excipients needed, and the pH is adjusted. Finally,
the preparation is sterile-filtered and dispensed into the final containers.
13.2.4 Optional additional steps used by some manufacturers
When performed following GMP and using validated fractionation protocols, the basic
methodologies described above for the manufacture of IgG, F(ab')2 and Fab antivenoms allow the
production of antivenoms of adequate purity, safety and efficacy. Nevertheless, some
manufacturers include additional steps to enhance product purity. The methodologies include
those described below.
13.2.4.1 Ion-exchange chromatography
Ion-exchange chromatography can be successfully used for antivenom purification based on
charge differential with the contaminants. Anion-exchange columns of DEAE or QAE gels or
membranes, such as quaternary ammonium cellulose microporous membranes, can be used at
neutral pH to adsorb protein contaminants (10, 70, 73). Alternatively, cation-exchange columns,
e.g. carboxymethyl or sulfopropyl gels, have been used for purification of IgG or F(ab')2
fragments (71). The column is equilibrated at acid pH, e.g. pH 4.5, to bind the antivenom,
whereas protein contaminants are eluted in the break-through.
Chromatographic procedures should be applied following GMP. Columns should be adequately
regenerated, sanitized, and stored to prolong their useful lifetime. The reproducibility of columns
over cycles should be validated. Measures to avoid batch to batch contamination should be in
place. Specific standard operating procedures should be developed and followed.
13.2.4.2 Affinity chromatography
Affinity chromatography using either immobilized venom or other ligands can be designed to
bind immunoglobulins or their fragments (74). However, columns usually deteriorate rather
rapidly, and meticulous care should be taken to wash, sanitize and store them under appropriate
conditions. Procedures should be followed to ensure that any substances leaching from the
columns do not affect the quality and safety of the product or else are completely removed
during downstream processing; this is especially critical in affinity chromatography using
immobilized venom. Affinity processes may affect recovery and high-affinity antibodies may be
lost and/or denatured owing to the harsh elution conditions needed to elute them from the
chromatographic material.
50
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Figure 6
Example of a fractionation process in which Fab fragments are prepared by papain
digestion and ammonium sulfate precipitation
51
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
13.2.4.3 Process improvement
Some manufacturers have introduced process improvements to enhance the quality or the yield
of antivenoms. These include the use of a depth filtration system combined with filter-aids to
facilitate filtration steps and improve antivenom recovery. In addition, other manufacturing steps
may be introduced to ensure inactivation or removal of infectious agents (see section 14).
13.2.5 Formulation
During formulation of antivenoms after diafiltration steps one should consider the addition of
salts to adjust the osmolality, addition of preservatives, other excipients, if needed for protein
stability, and the adjustment of pH.
In general, antivenoms are formulated at neutral pH (pH 7.0 ± 0.5) although some manufacturers
are exploring the feasibility of formulation at more acidic pH to improve stability and/or to
reduce aggregate formation.
Formulation at pH higher than 7.5 is not recommended, since the stability of immunoglobulins
and their fragments at alkaline pH may be poor, and the formation of aggregates may be
favoured.
13.2.6 Analysis of bulk product before dispensing
The biological, physical and chemical characteristics of the final bulk product should meet preestablished specifications before dispensing. Analysis may include tests required to demonstrate:
− the purity and potency of the product;
− the sterility;
− the compliance with the specifications for the aggregate content;
− the pyrogen limit and/or the bacterial endotoxin content; and
− the formulation.
When the product is formulated in liquid form, some of these tests (such as the potency assay)
may not need to be duplicated on the final container if the processing after the bulk preparation
has been validated and shown not to alter this activity.
The sterilization equipment and the integrity of the membrane should be guaranteed before
sterilization; moreover, the aseptic filling should be validated.
13.2.7 Dispensing and labelling of final product
Once compliance of the final bulk product with the quality control specifications is established,
the final product is bottled. For this, final glass containers (vials or ampoules) should be used.
General principles prevailing for the dispensing of parenteral medicinal products should be
applied. The dispensing should be performed in class A (59) clean room conditions, usually
under a laminar flow hood. The equipment used for dispensing should be calibrated beforehand
to ensure that the correct volume is delivered.
In the case of ampoules, the dispensing system should ensure an aseptic closure and the sealing
of the ampoule should prevent risk of protein denaturation due to heat. For vials, insertion of
rubber stoppers should be done inside this clean dispensing area. The quality of the rubber
stoppers should be such as to guarantee inertness and to prevent leaching. Thereafter, aluminium
seals should be placed on each vial in a clean area outside the class A area. Ampoules or vials
containing the final product should then be properly identified and stored in a quarantine area
52
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
maintained under proper storage conditions. Samples of the antivenoms should be sent to the
quality control laboratory for analysis.
When an antivenom complies with all the quality control tests established for the final product, it
should be properly labelled and identified. The vial or ampoule should be labelled with, at least,
the following information:
−
name of the product and of the producer;
−
animal species used to produce the antivenom;
−
batch number;
−
pharmaceutical presentation (liquid or freeze-dried);
−
volume content;
−
administration route;
−
specificity (venoms neutralized by the antivenom, including both the common and the
scientific name of the snake(s)1);
−
neutralizing potency;
−
storage conditions; and
−
expiry date.
Additional information may be requested by the national regulatory authorities. The package,
which is usually a cardboard box, in which the vials or ampoules are packed, should include the
same information as is given on the primary container. The package insert should include all the
information relating to the product, as established by national regulatory agencies, including:
− the neutralizing potency;
− the recommended dosage;
− reconstitution procedure, if lyophilized;
− the mode of administration (e.g. the dilution of antivenom in a carrier fluid such as
saline);
− the rate of administration;
− details on the symptoms and treatment of early and delayed adverse reactions;
− snake species against which the antivenom is effective;
− recommended storage conditions, and
− an indication that the product is for single use.
13.2.8 Use of preservatives
The addition of preservatives to prevent bacterial and fungal contamination should be kept to a
minimum during plasma storage and during fractionation. Their inclusion during the
manufacturing process should be clearly justified, and should never substitute for any aspect of
GMP. Preservatives can be considered in the final product, especially if it is manufactured in
liquid form, and most specifically preservatives are required for multiple-dose presentations.
Antimicrobial agents currently used in antivenom formulation include phenol and cresols. In
1
Special care should be taken considering the frequent changes in snake species taxonomy.
53
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
general, phenol concentration is adjusted to 2.5 g/l, and concentration of cresols should be less
than 3.5 g/l. The concentration of preservatives should be validated by each production
laboratory on the basis of assays to test their efficacy and keeping in mind that they may degrade
with time and cease to be effective. It is necessary to ascertain that any agent used has no
potential detrimental interaction with the active substance and excipients of antivenoms. Any
change in the formulation involving preservatives, or the elimination of preservatives from the
final product, requires a very careful risk–benefit assessment on various microbial safety aspects,
as well as a detailed validation procedure. Mercury-containing preservatives are not
recommended in antivenom manufacture. The volume of antivenom required for the treatment of
envenoming (in excess of 50 ml) might lead to an exposure to mercury far higher than the
amounts currently used for other biological preparations and levels at which they are toxic,
especially in young children, are not known (75, 76).
13.2.9 Freeze-drying
Antivenoms are available either as liquid or as freeze-dried preparations. Freeze-dried
antivenoms, which may usually be stored at a temperature not exceeding 25 °C, are generally
distributed to markets where the cold chain cannot be guaranteed, such as in many tropical
regions of the world. The absence of guarantee of a cold chain during distribution highlights the
need for manufacturers to demonstrate the stability of the antivenoms under the high
temperatures found in tropical climates.
Freeze-drying is a critical operation. Careful attention should be given to the rate of freezing as
well as to the protocol used for the primary and secondary drying cycles (77). The details of the
freeze-drying protocols are product-specific and should be adjusted according to the particular
formulation of each antivenom. Inadequate freeze-drying protocols may affect the
physicochemical quality of the product, inducing protein precipitation and denaturation, as well
as aggregate formation, and altering stability and reconstitution. Specific stabilizers, such as
sugars or polyols, aimed at protecting proteins from denaturation and aggregation, may be added
to the final formulation of the antivenom. Bulking agents, frequently used for some biological
products, are generally not required in the case of antivenoms owing to their relatively high
protein concentration; however they are sometimes used for high-titre monospecific antivenoms.
13.2.10 Inspection of final container
All the vials or ampoules of each batch of liquid antivenoms should be inspected, either visually,
or using a mechanical device. Any vial or ampoule presenting turbidity, abnormal coloration,
presence of particulate matter, or defects of the vial, stopper, or capsule should be discarded. In
the case of freeze-dried products, a representative sample of the whole batch should be dissolved
in the solvent and inspected as described. Turbidity can be assessed quantitatively by using a
turbidimeter.
13.2.11 Archive samples of antivenoms
In compliance with GMP, manufacturing laboratories should archive a number of vials of each
antivenom batch, under the recommended storage conditions, in an amount that would enable the
repetition of all quality control tests, when required.
54
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
13.3 Pharmacokinetic and pharmacodynamic properties of IgG, F(ab')2 and Fab
Owing to their different molecular mass, the pharmacokinetics of heterologous IgG molecules
(approximately 150 kDa) and F(ab')2 (approximately 100 kDa) and Fab (approximately 50 kDa)
fragments differ significantly. In envenomed patients, Fab fragments have the largest volume of
distribution and readily reach extravascular compartments. Fab fragments are, however, rapidly
eliminated, mainly by renal excretion, thus having a short elimination half-life (from 4–24 hours)
(78, 79). In contrast, F(ab')2 fragments and intact IgG molecules are not eliminated by the renal
route and therefore have a more prolonged elimination half-life (between 2 and 4 days) (11, 80,
81). Such different pharmacokinetic profiles have important pharmacodynamic implications, and
the selection of the ideal type of active substance in an antivenom should rely on a careful
analysis of the venom toxicokinetics and antivenom pharmacokinetics.
Another difference between low-molecular-mass fragments, such as Fab and those with a higher
molecular mass, such as F(ab')2 and IgG, is the number of paratopes of each molecule: Fab has
one antigen binding site while IgG and F(ab')2 each have two binding sites. Thus they will be
able to form large and stable complexes or precipitates with antigens carrying several epitopes,
while the former will form small, reversible non-precipitable complexes.
Ideally, the volume of distribution of an antivenom should be as similar as possible to the
volume of distribution of the main toxins in a particular venom; however, this is rarely the case.
In venoms composed of low-molecular-mass toxins, such as some elapid snake venoms, lowmolecular-mass neurotoxins are rapidly absorbed into the bloodstream and are rapidly distributed
to the extravascular spaces where toxin targets are located. Furthermore, low-molecular-mass
toxins are eliminated from the body in a few hours. In these cases, an antivenom of high
distribution volume that readily reaches extravascular spaces, such as as Fab, might be
convenient, although its action is then eliminated within a few hours. It should be noted,
however, that a number of elapid venoms contain some high-molecular-mass toxins of great
clinical significance, such as procoagulants and pre-synaptic neurotoxins.
In contrast, in the case of viperid snake venoms and other venoms made up of toxins of larger
molecular mass, including a number of elapid venoms, many of which act intravascularly to
provoke bleeding and coagulopathy, the situation is different. The time required for toxins to
distribute to extravascular spaces is longer than in the case of low-molecular-mass neurotoxins,
and the targets of some of these toxins are present in the vascular compartment. In addition, the
toxins of viperid venoms have a long half-life in vivo and can remain in the body for several
days (82, 83). In this case, an antivenom made by Fab fragments neutralizes the toxins that reach
the circulation but, after a certain time has elapsed, the Fab fragments are eliminated and nonneutralized toxins reach the circulation, thus giving rise to the well-known phenomenon of
recurrent envenoming, i.e. the reappearance of signs and symptoms of envenoming at later time
intervals after an initial control of envenoming. This situation demands repeated administration
of antivenom to maintain therapeutic levels of Fab in the circulation (84). Therefore, in such
envenomings, antivenoms made of IgG or F(ab')2 may be more appropriate because of their
longer elimination half-lives. Moreover, it has been proposed that formation of venom–
antivenom complexes in the circulation results in the redistribution of venom components from
the extravascular space to the blood compartment, where they are bound and neutralized by
circulating antivenom, provided that the dose of antivenom is sufficient (85, 86). Consequently,
the maintenance of a high concentration of specific antivenom antibodies in the circulation for
many hours is required for complete neutralization of toxins reaching the bloodstream during
both early and late phases of envenoming (redistribution of toxins) present in the extravascular
space. In conclusion, IgG and F(ab')2 antivenoms have a pharmacokinetic profile that makes
them more effective in many types of snakebite envenoming.
55
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
13.4 Main recommendations
•
Antivenoms should be manufactured using fractionation procedures that are well
established, validated, and shown to yield products with proven safety and efficacy.
Fractionation processes used for the manufacture of antivenoms should adhere to
the principles of GMP for parenteral medicinal products.
•
Antivenoms can be comprised of intact IgG molecules, F(ab')2 fragments or Fab
fragments. Intact IgG antivenoms are mainly produced by caprylic acid
precipitation of non-IgG plasma proteins, leaving a highly purified IgG preparation
in the supernatant or filtrate.
•
F(ab')2 fragment antivenoms are produced by pepsin digestion of plasma proteins,
at acidic pH, usually followed by F(ab')2 purification by salting out with ammonium
sulfate solutions or by caprylic acid precipitation. Fab monovalent fragments are
obtained by papain digestion of IgG at neutral pH.
•
Further to ultrafiltration to remove low-molecular-mass contaminants,
preparations are formulated, sterilized by filtration and dispensed in the final
containers. Formulations of antivenoms may include preservative agents.
•
Antivenoms can be presented as liquid or freeze-dried preparations. Freeze-drying
of antivenoms should be performed in conditions that ensure no denaturation of
proteins and no formation of protein aggregates.
•
IgG, F(ab')2 and Fab antivenoms exhibit different pharmacokinetic profiles: Fab
fragments have a larger distribution volume and a much shorter elimination halflife. Thus, for viperid envenomings, IgG or F(ab')2 antivenoms have a more suitable
pharmacokinetic profile, whereas Fab fragments may be useful for the
neutralization of venoms rich in low-molecular-mass neurotoxins which are rapidly
distributed to the tissues. However, in general terms, IgG and F(ab')2 antivenoms
have shown a better pharmacokinetic profile than Fab antivenoms.
14 CONTROL OF INFECTIOUS RISKS
14.1 Background
The viral safety of any biological product results from a combination of measures to ensure a
minimal risk of viral contamination in the starting material (e.g. plasma), together with steps to
inactivate or remove potential contaminating viruses during processing.
There are currently several recognized complementary approaches used for virus risk reduction
for biological products. These are:
− minimizing the potential initial virus content by implementing a quality system for the
production of the starting material;
− contribution of the manufacturing processes to inactivating and/or removing residual
viruses during manufacture of the biological product; such a contribution can be inherent
to the existing production technology or may result from the introduction of dedicated
viral reduction steps;
− adhesion to GMP at all steps of the manufacturing process;
− appropriate and timely response to any infectious events recognized during the clinical
use of the product.
56
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Production steps to inactivate and/or remove viruses have long been shown to play a powerful
role in ensuring safety of biologicals (60). Similarly, keeping to a minimum the potential viral
load at the stage of the plasma pool, through appropriate epidemiological surveillance and health
control of the donor animals, is also an important safety measure (see section 10).
Based on experience with human plasma products, a production process for antivenoms that
includes two robust steps for viral reduction (comprising preferably at least one viral inactivation
step) should provide a satisfactory level of viral safety. However, it should be kept in mind that
non-enveloped viruses are more difficult to inactivate or remove than lipid-enveloped viruses.
14.2 Risk of viral contamination of the starting plasma
The main structural characteristics of viruses reported to possibly infect horses, sheep and goats
are presented in Tables 7 and 8. They include viruses with a DNA or RNA genome, with and
without a lipid envelope, and varying widely in size (22 to 300 nm).
A few of these viruses have been identified as possibly present, at least at some stages of the
infection cycle, in the blood, or are considered as being pathogenic to humans. Special attention
should be paid to these viruses.
14.3 Viral validation of manufacturing processes
Understanding how much a manufacturing process may contribute to the viral safety of
antivenoms is fundamental to both manufacturers and regulators. Such an understanding can
only be achieved by viral validation studies. These studies are complex and require wellestablished virology laboratory infrastructure and cell culture methodologies. They are usually
carried out by specialized laboratories, outside the manufacturing facilities.
The principles guiding such studies have been described in WHO Guidelines (60) and are
summarized below.
14.3.1 Down-scale experiments
The contribution of manufacturing processes towards inactivation and/or removal of potential
viral contamination should be demonstrated. For this purpose, viral validation studies should be
performed using at least three viruses exhibiting different structural characteristics. The
antivenom manufacturer should first identify the steps that, based on the existing literature, are
likely to remove or inactivate viruses and, then, provide evidence and quantitative assessment of
the extent of virus reduction achieved for the specific process evaluated.
Validation should be done by down-scale experiments. The accuracy of the down-scale process
should be assessed by comparing the characteristics of the starting intermediate and the fraction
resulting from that step, for both the laboratory and the production scales. Selected physical
factors (e.g. temperature, stirring or filtration conditions) and chemical factors (e.g. pH or
concentration of precipitating agents such as caprylic acid) should be as close as possible to
those used at manufacturing scale.
57
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Table 7
Viruses identified in horses (62, 87)
Virus
Size
(nm)
Family
Genome
Presence in
blood
reporteda
Classified as
pathogenic to
humans (62)
Lipid-enveloped viruses
Borna virus
b
Bornaviridae
70–130
ss-RNA
Yes
Equine arteritis virus
Arteriviridae
50–60
ss-RNA
Equine encephalitis virus,
Eastern and Western
Togaviridae
40–70
ss-RNA
Equine coronavirus
Coronaviridae
75–160
ss-RNA
Equine foamy virus
Retroviridae
80–100
ss-RNA
Equine herpes virus 1–5
Herpesviridae
125–150
Ds-DNA
Equine infectious anaemia virus
Lentiviridae
80–100
ss-RNA
Equine influenza virus
Orthomyxoviridae
80–120
ss-RNA
Yes
Equine morbillivirus (Hendra
virus)
Paramyxoviridae
150
ss-RNA
Yes
Japanese encephalitis virus
Flaviviridae
40–70
ss-RNA
Yes
Nipah virus
Paramyxoviridae
150–300
ss-RNA
Yes
Rabies virus
Rhabdoviridae
75–180
ss-RNA
Yes
Salem virus
Paramyxoviridae
150–300
ss-RNA
St Louis encephalitis virus
Flaviviridae
40–70
ss-RNA
Venezuelan equine encephalitis
virus
Togaviridae
40–70
ss-RNA
Yes
Yes
Vesicular stomatitis virus
Rhabdoviridae
50–80
ss-RNA
Yes
Yes
West Nile virus
Flaviviridae
40–70
ss-RNA
Yes
Yes
Yes
Yes
Yes
Yes
Yes
Non-lipid enveloped viruses
Equine encephalosis viruses
Reoviridae
Equine rhinitis A and B viruses
Equine rotavirus
a
b
58
80
Ds-RNA
Picornaviridae
22–30
ss-RNA
Reoviridae
60–80
Ds-RNA
Absence of a report does not mean that the virus may not be found in the blood at certain stages of the cycle of
infection.
Recent studies have suggested that Borna virus is non-pathogenic to humans (88).
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Table 8
Viruses identified in sheep and goats (62)
Virus
Family
Size (nm)
Genome
Classified as
Reported
presence in pathogenic to
humans (62)
blooda
Lipid-enveloped viruses
Adenovirus
Adenoviridae
80–110
ds-DNA
Akabane virus
Bunyaviridae
80–120
ss-RNA
Bluetongue virus
Reoviridae
80
ds-RNA
Border disease virus
Flaviviridae
40–70
ss-RNA
Borna virusb
Bornaviridae
70–130
ss-RNA
Bovine herpes virus types 1, 2, 4
Herpesviridae
120–200
ds-DNA
Bovine viral diarrhoea virus
Togaviridae
40–60
ss-RNA
Loiping ill virus
Flaviviridae
40–50
ss-RNA
Nairobi sheep disease
Bunyaviridae
80–120
ss-RNA
Ovine/bovine papillomavirus
Papillomaviridae
40–55
ds-DNA
Ovine herpes virus 2
Herpesviridae
120–200
ds-DNA
Parainfluenza virus type 3
Paramyxoviridae
150–300
ss-RNA
Peste des petits ruminants
(Morbillivirus)
Paramyxoviridae
150–300
ss-RNA
Poxviruses (Parapox, Capripox,
Cowpox)
Poxviridae
140–260
ds-DNA
Respiratory syncytial virus
Paramyxoviridae
150–300
ss-RNA
80–100
ss-RNA
Retroviruses (Caprine arthritis
encephalitis virus, Maedi-Visna virus,
Jaagsiekte virus, Bovine leukaemia
virus)
Retroviridae
Rift Valley fever complex
Bunyaviridae
80–120
ss-RNA
Ross river virus
Togaviridae
70
ss-RNA
Rotavirus
Reoviridae
80
ds-RNA
Tick-borne encephalitis virus
Flaviviridae
40–50
ss-RNA
Vesicular stomatitis virus
Rhabdoviridae
50–380
ss-RNA
Wesselbron virus
Flaviviridae
40–50
ss-RNA
Yes
Yes
Yes
Yes
Yes
Yes
Yes
Yes
Yes
Yes
Non-lipid enveloped viruses
Epizootic haemorrhagic disease virus
Reoviridae
Foot and mouth disease virus
Reovirus 1-3
a
b
80
ds-RNA
Picornaviridae
27–30
ss-RNA
Reoviridae
60–80
ds-RNA
Yes
Absence of report does not mean that the virus may not be found in the blood at certain stages of the cycle of
infection.
Recent studies have suggested that Borna virus is non-pathogenic to humans (88).
59
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Once the step is accurately modelled, the antivenom fraction derived from the fractionation
process just prior to the step being evaluated (e.g. the starting plasma to be subjected to a low pH
treatment, or to caprylic acid precipitation, or a F(ab')2 fragment fraction to be subjected to an
ammonium sulfate-heat treatment) should be spiked with one of the model viruses selected. Viral
infectivity, most often determined using cell culture assays (less frequently animal models),
should be quantified before (e.g. prior to pH adjustment and addition of pepsin) and immediately
after (e.g. following low pH adjustment and incubation at that pH for a known period of time in
the presence of pepsin) the steps evaluated to determine the viral clearance achieved. The results
are conventionally expressed as logarithm (log) of the reduction in infectivity that is observed.
Total infectivity or viral load is calculated as the infectious titre (infectious units per ml)
multiplied by the volume. For a viral inactivation step, it is highly recommended that the kinetics
of the virus kill be evaluated. Such inactivation kinetics of the infectivity provides an important
indication of the virucidal potential of the step and enables comparison of the data obtained to
those from published studies.
Typically, a viral reduction of 4 logs or more is considered to represent an effective and reliable
viral safety step.
Establishing the relative insensitivity of a manufacturing step to changes or deviatons in process
conditions is also important to evaluate its robustness, in addition to adding to the level of
understanding of its contribution to the overall viral safety of the preparation. This can be
achieved by validating the same step using a range of conditions deviating from those used in
production (such as an upper pH limit applied to a pepsin digestion or to a caprylic acid
precipitation step).
Virus validation studies are subject to a number of limitations (60), which should be considered
when interpreting the results.
14.3.2 Selection of viruses for the validation of antivenom production processes
Viruses selected for viral validation studies should resemble as closely as possible those which
may be present in the starting animal plasma material (Tables 7 and 8). When possible, viruses
known to potentially contaminate animal plasma (called “relevant viruses”) should be used.
Table 9 gives examples of a few viruses that have been used for the validation of animal-derived
immunoglobulins. Vesicular stomatitis virus (VSV) and West Nile virus (WNV) are relevant
lipid-enveloped horse plasma-borne viruses. Bovine viral diarrhoea virus (BVDV), a lipidenveloped flavivirus, can be used as a model for West Nile virus (WNV) and for the Eastern,
Western, and Venezuelan equine encephalitis togaviruses. Pseudorabies virus (PRV) is a lipidenveloped virus that can serve as a model for pathogenic equine herpesvirus.
Encephalomyocarditis virus (EMCV), a picornavirus, can serve as a model for non-lipidenveloped viruses. Porcine parvovirus can also be selected as a model for small resistant nonlipid-enveloped viruses or as a relevant virus when pepsin of porcine origin is used in the
manufacture of F(ab')2 fragments.
This list is not exhaustive and other model viruses can be used for validation studies of animalderived antivenoms, in particular taking into account the characteristics of the viruses that may
be present in the animal species used to generate antivenoms.
60
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Table 9
Examples of laboratory model viruses that can be used for validation studies of horsederived antivenoms
Virus
Family
LipidSize (nm)
enveloped
Genome
Resistance Model for
Parvoviridae
No
18–26
SS-DNA
High
Relevant virus (when
pepsin of porcine
origin is used)
Togaviridae
Yes
40–60
ss-RNA
Low
Eastern, Western and
Venezuelan equine
encephalitis virus
Parainfluenza virus
Paramyxoviri
dae
Yes
100–200
ss-RNA
Low
Hendra virus; Nipah
virus; Salem virus
Poliovirus;
Encephalomyocarditi
s virus; Hepatitis A
virus
Picornaviridae
No
25–30
ss-RNA
Mediumhigh
Equine rotavirus
Pseudorabies virus
Herpes
Yes
100–200
Ds-DNA
Medium
Equine herpes virus
Reovirus type 3
Reoviridae
No
60–80
Ds-RNA
Medium
Equine encephalosis
virus
Sindbis virus
Togaviridae
Yes
60–70
ss-RNA
Low
Eastern, Western and
Venezuelan equine
encephalitis virus
Vesicular stomatitis
virus
Rhabdoviridae
Yes
50–200
ss-RNA
Low
Relevant virus
Low
Relevant virus and
model for Eastern
equine encephalitis
virus
Animal Parvovirus
(e.g. porcine)
Bovine virus
Diarrhoea virus
West Nile virus
Flaviridae
Yes
40–70
ss-RNA
14.4 Viral validation studies of antivenom immunoglobulins
There is no documented case of transmission of zoonotic infections, including viral diseases, by
antivenom immunoglobulins, or any other animal-derived immunoglobulins. Absence of reports
of viral transmission may result from a lack of long-term surveillance of the patients receiving
antivenoms. Alternatively, this may reveal that current processes for the manufacturing of
antivenoms include processing steps that contribute to the viral safety.
Among the various processing steps used in the production of antivenoms, caprylic acid and low
pH treatments are known to contribute to safety against lipid-enveloped viruses. This
information is based on well-established experience in the fractionation of human plasma with a
production step comprising caprylic acid (89–91) or low pH treatment (60, 92–94).
Although information is still limited, there is now growing evidence that similar steps used in the
production of animal derived immunoglobulins may also inactivate or remove viruses. In
addition, some manufacturers have implemented dedicated viral reduction procedures.
61
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
14.4.1 Caprylic acid treatment
The conditions used for caprylic acid treatment of antivenoms (87, 64) and of human
immunoglobulins (89–91) are similar, in particular the pH range, duration of treatment,
temperature, and the caprylic acid/protein ratio, as summarized in Table 10.
Table 10
Comparison of conditions for caprylic acid treatment used for human immunoglobulin
preparations and antivenoms (87)
Temperature
Duration
( °C)
(hr)
5.5
22
1
15
4.8
20
1
25
20
5.0
20
1
60–90
50
5.5–5.8
18–22
1
Protein
concentration (g/l)
Caprilic acid (g/kg
solution)
pH
Human IgG
35
7.45
Human IgMenriched
43
Human IgM
Antivenoms
Product
14.4.1.1 Validation studies with human immunoglobulins
Unsaturated fatty acids, most specifically caprylic acid, have long been known to have the
capacity to inactivate lipid-enveloped viruses in human plasma protein fractions (95, 96). The
non-ionized form of caprylic acid is thought to disrupt the lipid bilayer and membrane-associated
proteins of enveloped viruses. Utilizing the dissociation reaction and varying the concentration
of the ionized form of caprylate, a specific amount of the non-ionized form of caprylate can be
maintained over a wide pH range.
The robustness of a caprylic acid treatment applied to human immunoglobulin G (IgG), human
immunoglobulin M (IgM) and IgM-enriched preparations has been investigated using various
enveloped viruses (human immunodeficiency virus (HIV), BVDV, Sindbis virus and
Pseudorabies) (89). Under the conditions applied during manufacture, caprylic acid leads to
robust inactivation of lipid-enveloped viruses; pH is a particularly critical parameter and should
be less than 6.
Another investigation studied the viral reduction achieved during treatment by caprilate of a
human IgG product (90). At pH 5.1, at 23 °C, and in the presence of 9 mM caprylate, ≥ 4.7 and ≥
4.2 log of HIV and PRV, respectively, were inactivated during the 1-hour treatment, but only 1.5
log for BVDV was inactivated. At 12 mM caprylate, ≥ 4.4 log of BVDV were inactivated within
this time period. At pH 5.1, 24 °C, and 19 mM caprylate, and pH 5.1, 24 °C, and 12 mM
caprylate, complete inactivation of BVDV and of HIV and PRV was achieved in less than 3 min.
14.4.1.2 Validation studies with antivenom immunoglobulins
Virus inactivation studies have been carried out on a F(ab')2 fraction obtained from pepsin
digested plasma subjected to ammonium sulfate precipitation. The F(ab')2 fraction was subjected
to precipitation by drop-wise addition of caprylic acid to 0.5% (final concentration) and the
mixture was maintained under vigorous stirring for 1 hour at 18 °C. Rapid and complete
reduction of BVDV, PRV and VSV (> 6.6 log10, > 6.6 log10, and > 7.0 log10, respectively) was
observed. No significant reduction (0.7 log10) of the non-enveloped EMCV (97) was observed.
In another process used to prepare equine immunoglobulins, in which serum is thawed at 4 °C,
subjected to heating at 56° C for 90 min, brought to 20 ± 5 °C, adjusted to pH 5.5 and subjected
to 5% caprylic acid treatment for 1 hour, fast reduction of infectivity of > 4.32 and > 4.65 log10
62
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
was found for PRV and BVDV, respectively. The caprylic acid step was confirmed to have
limited impact on the infectivity of EMCV and Minute virus of mice (MVM) non-lipid
enveloped viruses (98). Data suggest that significant reduction in the infectivity of lipidenveloped viruses can be obtained during caprylic acid treatment of antivenoms. The reduction
of viral infectivity may result from both viral inactivation and partitioning during the
precipitation step. No significant inactivation of non-enveloped viruses is expected.
14.4.1.3 Recommended actions
Further studies of the viral reduction achieved during caprylic acid treatment of antivenoms are
recommended; in particular, robustness studies to define the impact on process variations should
also be performed.
14.4.2 Acid pH treatment
The conditions used for low pH treatment of equine derived antivenom immunoglobulins and of
human immunoglobulins are summarized in Table 11.
Table 11
Typical conditions for acid pH treatment of human IgG preparations and equine
antivenoms (87)
Product
Protein concentration (g/l)
pH
Temperature ( °C)
Duration (hrs)
Human IgG
40–60
4
30–37
20–30
Antivenoms
60–90
3.1–3.3
30–37
0.6–24
14.4.2.1 Validation studies with human immunoglobulins
Many studies have demonstrated that the low pH 4 treatment used in the manufacture of human
intravenous IgG has the capacity to inactivate lipid-enveloped viruses (92–94). The rate and
extent of inactivation may differ depending upon the virus. Inactivation is temperaturedependent, and is influenced by the formulation of the IgG solution. Pepsin is sometimes added
in traces (to reduce anticomplementary activity and content of aggregates) but, at this low
concentration, contributes little to virus kill (60). Most non-lipid enveloped viruses are resistant
to acid pH treatment.
14.4.2.2 Virus inactivation studies performed with antivenom immunoglobulins
As described in section 13, peptic cleavage of horse plasma IgG at pH 3.0–3.3 for 60 min is a
common procedure for the preparation of F(ab')2. More than 4 logs of inactivation of WNV and
of Sindbis has been found when horse plasma was subjected to peptic digestion at pH 3.2 for 60
min (99). WNV was very sensitive whether pepsin was added or not, whereas the rate and extent
of inactivation of Sindbis was higher in the presence of pepsin. This suggests that pH 3.2 alone
inactivates WNV, while other phenomena involving the action of pepsin contribute to Sindbis
inactivation at low pH.
Confirmation of the significant inactivation of lipid-enveloped viruses during peptic cleavage of
plasma at pH 3.2 was obtained by another group (97). In this process, plasma is diluted with two
volumes of saline, pH is adjusted to 3.3, and pepsin is added to a final concentration of 1g/l. The
mixture is incubated at pH 3.3 for 1 hour. Inactivation of PRV > 5.1 log10 occurred in less than 6
minutes and > 7.0 log10 in 60 min. There was > 3.1 log10 and > 4.5 log10 inactivation of VSV after
6 and 20 min, respectively. The reduction of infectivity of BVDV was less: 1.7 log10 after 60
min. Inactivation of EMC, a non-enveloped virus, was relatively slow but reached between 2.5
63
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
and 5.7 log10 after 60 min of pepsin incubation. This showed that reduction of infectivity of at
least some non-lipid enveloped viruses may take place during peptic digestion of diluted horse
plasma. This does not mean, however, that other non-lipid enveloped viruses would be
inactivated to the same extent under such conditions.
14.4.2.3 Recommended actions
Manufacturers of F(ab')2 antivenoms are encouraged to validate the pepsin digestion process
since virus inactivation is likely to be influenced by pH, time, temperature, pepsin content, and
protein content. Robustness studies to define the impact on process variations are also
recommended.
14.4.3 Filtration steps
Other steps used in antivenom production may contribute to viral safety through non-specific
viral removal. The virus removal capacity of two depth-filtration steps performed in the presence
of filter aids and used in the production of equine derived immunoglobulins by ammonium
sulfate precipitation of pepsin-digested IgG has been evaluated (100). Clearance factors of 5.7
and 4.0 log10 have been found for two lipid-enveloped viruses (infectious bovine rhinotraceheitis
virus and canine distemper virus, respectively) and of 5.3 and 4.2 log10 for two non-lipid
enveloped viruses (canine adenovirus virus and poliovirus type I, respectively). However, it
should be kept in mind that viral reductions obtained by non-dedicated removal steps are usually
regarded as less robust than those resulting from dedicated viral inactivation or removal steps
(60).
14.4.4 Validation of dedicated viral reduction treatments
14.4.4.1 Pasteurization
Pasteurization is defined as the treatment of a liquid protein fraction for 10 hours, usually at
60 °C. It is a well-established viral inactivation treatment of human plasma products, such as
immunoglobulin G (60). It is being used in the production process of a few equine-derived
immunoglobulins (10).
Validation studies showed that heating a purified equine immunoglobulin at 58 °C ± 0.1 °C
without stabilizers inactivates ≥ 4.8 log10 of PRV and ≥ 4.3 log10 of BVDV in less than 30 min,
and > 4.7 log of EMCV in less than 1 hour. In contrast, infectivity of MVM, a non-enveloped
virus, was still detected after 9 h and 30 min of treatment; only 1.59 log10 were inactivated (98).
14.4.4.2 Nanofiltration
Nanofiltration is a technique of filtration specifically designed to remove viruses, based on size,
while permitting flow-through of the desired protein (101). Effective viral removal requires, in
principle, that the pore size of the filter be smaller than the effective diameter of the virus
particles.
14.4.5 Other viral inactivation treatments currently not used in antivenom manufacture
Other methods of viral inactivation have been developed to ensure the viral safety of biological
products. These include, in particular, a treatment with a combination of an organic solvent (trin-butyl phosphate or TnBP) at concentrations between 0.3 and 1%, and detergents such as Triton
X-100 or Tween 80, also at concentrations generally between 0.3% and 1%. Such solvent–
detergent (S/D) procedures have proven to be very efficient and robust in the inactivation of
lipid-enveloped viruses in human plasma products (60). However, use of this method for
antivenoms has not been reported.
64
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Implementation of dedicated viral inactivation treatments, such as S/D or other methods, should
be encouraged for processes which, based on risk assessment, would offer an insufficient margin
of viral safety. Process changes associated with the introduction of new viral reduction steps, and
the subsequent removal of any toxic compounds needed for viral inactivation, should be
demonstrated not to affect the quality and stability of antivenoms, and most particularly the
neutralization efficacy of venoms. Preclinical assessment of the possible impact of newly
introduced viral inactivation treatments should be mandatory.
14.4.6 Possible contribution of phenol and cresols
The anti-bacterial agents, phenol or cresols, and more rarely formaldehyde, are added, by most
manufacturers to the starting plasma donations as well as to the final liquid antivenom
preparations, at a maximum final concentration of 0.25–0.35%. Compounds like phenol are
known to be very lipid-soluble and lipophilic.
Performing validations of the potential virucidal effect of antimicrobial agents as added to the
starting hyperimmune plasma and to the final antivenom preparations is encouraged. More
information is needed on the potential impact of these antimicrobial agents on the viral safety of
antivenoms.
14.5 Production-scale implementation of process steps contributing to viral safety
As there is increasing, although preliminary, evidence that at least some of the existing steps in
the production of antivenoms contribute to viral reduction, it is already recommended that
specific care should be taken to ensure their appropriate industrial implementation so as not to
compromise any possible benefits they provide on viral safety.
Measures should therefore be taken to ensure that such steps are correctly carried out in a
manufacturing environment and that cross-contamination and downstream-contamination are
avoided. Such important aspects of product safety have been highlighted recently in WHO
Guidelines (60) and should also be taken into consideration for large-scale manufacture of
antivenoms. Specific attention should be given to:
− Process design and layout, in particular the production floor area needed to carry out
such treatment safely, the justification for creating a safety zone to avoid risk of downstream contamination, and the procedures used for cleaning and sanitization of the
equipment to avoid batch-to-batch cross-contamination.
− Equipment specifications, having in mind the potential contribution to viral reduction.
For instance, vessels used for low pH incubation or caprylic acid treatment should be
fully enclosed and temperature-controlled. There should be no “dead points” where the
temperature defined in the specification or the homogeneity of mixing cannot be
ensured. A poor equipment design could compromise the viral safety potentially
afforded by a given production step.
− Qualification and validation: should verify that the equipment conforms to predefined
technical specifications and relevant GMP.
− Process implementation: production steps contributing to viral safety such as low pH
treatment and caprylic acid treatments could be implemented in two stages performed in
two distinct enclosed tanks. Care should be taken to ensure complete process segregation
before and after the completion of these treatments to avoid risks of downstream
contamination.
65
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
− Process control: is a critical part of the manufacturing process since completion of viral
inactivation and removal cannot be guaranteed by testing the final product. Samples
should be taken to confirm that the process conditions of claimed inactivation steps meet
the specified limits (e.g. for pH, concentration of stabilizers and concentration of virus
inactivating agent, such as caprylate). When this is technically feasible and intermediates
are stable, samples can be kept frozen for possible additional analysis prior to the release
of the batch. It is the responsibility of the quality assurance department to ensure that the
execution of steps contributing to virus inactivation and removal in a production setting
conforms to the conditions that contribute to such virus reduction.
− Standard operating procedures: steps contributing to viral reduction should be described
in approved standard operating procedures. These should contain critical process limits
for the viral inactivation and removal methods.
− Role of the quality assurance department: because of the critical nature of the viral
inactivation and removal steps, quality assurance personnel should review and approve
the recorded conditions for viral inactivation and removal while the batch is being
processed; i.e. not just as part of the final overall review of the batch file.
14.6 Transmissible spongiform encephalopathy
Transmissible Spongiform Encephalopathy (TSE) has not been identified in any equine species.
There has been no case of transmission of TSE linked to antivenoms or other equine-derived
blood products.
Of particular concern, however, is the fact that TSEs include scrapie in sheep and goats,
ruminant species used, although less frequently than the horse, in the manufacture of
antivenoms. Scrapie is a disease similar to bovine spongiform encephalopathy (BSE or “madcow” disease), but is not known to infect humans. However, the blood of sheep with
experimental BSE or natural scrapie can be infectious, and scrapie has been experimentally
transmitted to monkeys. Because the infectious agents of scrapie and BSE behave similarly in
sheep and goats, the use of the blood of small ruminants should either be avoided in preparing
biologicals or be selected very carefully from sources known to be free of TSEs. The recent
findings of disease-associated proteins in muscle tissue of sheep with scrapie and the recognition
of BSE itself in a goat, reinforce the need for manufacturers of biologicals, including antivenoms,
to maintain the precautionary safety measures recommended in the WHO guidelines on TSE
tissue infectivity (54).
According to these recommendations, the use of tissues or body fluids of ruminant origin should
be avoided in the preparation of biological and pharmaceutical products. When sheep materials
must be used, they should therefore be obtained from animals assessed to have negligible risk of
scrapie. The feed of animals used for production of hyperimmune plasma should be free of
ruminant-derived material. Documented surveillance records should be available.
The infectious TSE agents are associated with misfolded, abnormal, prion protein (PrPTSE). It is
not yet known whether manufacturing processes used to produce antivenoms from sheep plasma
include steps that can contribute to the removal of PrPTSE. Experimental prion clearance studies,
based on spiking experiments, can be performed to assess the capacity of the process to remove
PrPTSE and infectivity. However, there is still uncertainty about the validity of such experimental
studies since PrPTSE has not been detected in blood and plasma and the properties of infectivity
in blood are not well understood.
66
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
14.7 Main recommendations
•
The viral safety of antivenoms results from a combination of measures:
− to ensure satisfactory health status of the animals;
− to reduce the risk of contamination in the starting material;
− to ensure the contribution of the manufacturing process towards inactivation
and/or removal of viruses; and
− to ensure compliance with GMP all along the chain of production.
•
Manufacturing processes should include at least one, and, preferably, two steps
contributing to robust viral reduction. A virus inactivation step that can be easily
monitored is usually preferred to other means of viral reduction, such as nonspecific removal.
•
Manufacturers are encouraged to evaluate and validate the capacity of their current
manufacturing processes (in particular low pH pepsin digestion, caprylic acid
treatment, ammonium sulfate or heat precipitation, and possibly other steps) to
inactivate or remove viruses. These studies should be done following existing
international guidelines and using relevant and/or model viruses that are
representative of the viruses that could affect the animals used for the production of
the antivenom immunoglobulins.
•
The removal of antimicrobial agents from the final formulation of antivenoms
should be carefully weighed against the potential benefits these agents may have on
the viral safety.
•
Should the viral reduction processes used be found to be insufficient to ensure a
margin of safety, the introduction of dedicated viral reduction methods should be
considered. The impact of such process changes on product efficacy and safety
should be carefully analysed in vitro as well as in preclinical studies before
performing clinical evaluations in humans.
•
Great attention should be paid to the production-scale implementation of all steps
contributing to viral safety to ensure a consistent and reproducible batch-to-batch
viral reduction and an absence of risks of cross-contamination and downstream recontamination that would jeopardize the viral safety of the product.
•
When sheeps are to be used for the production of plasma, the animals should be
obtained from sources assessed to have negligible risk of scrapie.
67
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
15
QUALITY CONTROL OF ANTIVENOMS
The quality control of the final product is a key element in the quality assurance of antivenoms.
Quality control tests should be performed by the manufacturer or under its responsibility before
the product is released. In addition, relevant analyses should be performed on any intermediate
steps of the manufacturing protocol as part of the in-process quality control.
The results obtained should meet the specifications approved for each antivenom product or its
intermediates, and are part of the batch record. For a liquid preparation, some quality control
tests, such as the potency test or the detection of residual reagents used during fractionation, can
be performed on the final bulk and may not need to be repeated on the final product if the
processing after the bulk preparation has been validated and shown not to have any impact. The
quality control of the final product in antivenoms includes the tests described below.
15.1 Tests
15.1.1 Appearance
The appearance of the product (e.g. colour of the liquid, appearance of the powder) should
comply with the description in the marketing dossier.
15.1.2 Solubility (freeze-dried preparations)
The time from the addition of solvent to the complete dissolution of freeze-dried antivenom,
under gentle mixing, should be determined. Antivenoms should be completely dissolved within
10 minutes at room temperature. The solution should not be cloudy. Shaking of the container
should be avoided to prevent the formation of foam.
15.1.3 Extractable volume
The volume of product extractable from the container should be in compliance with that
indicated on the label.
15.1.4 Venom-neutralizing potency test
This test determines the effectiveness of the antivenom to neutralize the overall toxic activity of
the snake venom(s) against which the antivenom is designed to act. The first part of the test, to
determine the lethal activity of the venom, is called the median lethal dose (LD50) assay and
usually uses mice of a defined weight range (e.g. 18–20 g). For new venoms whose LD50 is
unknown, it is recommended that a range dose-finding study, using one mouse per venom dose,
is performed to avoid using excessive numbers of animals. Some producers use other test
animals, such as guinea-pigs. While weights will clearly vary between animal species, the
following principles, specified for mice, will still apply to these alternative test animals. It should
be borne in mind that there are variations in the susceptibility of various strains of mice to the
lethal effect of venoms.
LD50 range-finding test: Various venom doses are prepared using saline solution as
diluent, and aliquots of a precise volume (0.2–0.5 ml) of each dose are injected, using one mouse
per dose, by the intravenous route, in the tail vein (or, alternatively, by the intraperitoneal route
(using injection volumes of 0.5 ml)). Deaths are recorded at 24 hours (intravenous test) or at 48
hours (intraperitoneal test). On the basis of this preliminary dose-finding experiment, a range of
venom doses causing 0% to 100% lethality is established and thus narrows the range of venom
doses required to formally estimate the toxic activity of the venom.
68
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
The median lethal dose (LD50) assay: Groups of 5–6 mice of a defined weight range are
injected intravenously, in the tail vein, with a precise volume (0.2–0.5 ml) of solutions of varying
doses of venom dissolved in sterile saline solution. A minimum of 5 mice is the smallest number
recommended for obtaining a statistically significant result. In some laboratories the LD50 is
estimated by the intraperitoneal route using an injection volume of 0.5 ml. Deaths are recorded at
24 hours (for assays involving intravenous injections) or at 48 hours (when intraperitoneal
injections are used), and LD50 is estimated by Probit analysis (102), Spearman-Karber (8) or
alternative procedures (such as non-parametric methods). One venom LD50 is defined as the
minimal amount of venom causing death in 50% of the mice injected. The test to assess the
neutralizing potency of an antivenom is called the median effective dose (ED50) assay. For a new
antivenom, it is recommended that a preliminary range dose-finding procedure is performed,
using one mouse per antivenom dose.
ED50 range-finding test: The selected multiple of the venom LD50 (3–6 LD50) is mixed
with different doses of antivenom and incubated at 37oC for 30 minutes and each mixture
injected into a single mouse. This preliminary test should establish a range of antivenom
volumes that result in 100% survival and 100% death of the injected mice and thus narrows
down the range of doses required for the formal ED(50) test.
The median effective dose (ED50) assay: This test involves the incubation of a fixed
amount of venom (“challenge dose”, usually corresponding to 3–6 LD50), with various volumes
of the antivenom adjusted to a constant final volume with saline solution (53, 103, 104). The
mixtures are incubated for 30 minutes at 37 °C, and then aliquots of a precise volume (0.2–0.5 ml)
of each mixture are injected into groups of generally 5 or 61 mice of a defined weight range by
the intravenous route, using the tail vein. A control group injected with a mixture of the venom
“challenge dose” with saline solution alone (no antivenom) should be included to confirm that
the venom “challenge dose” induces 100% lethality. When the test is performed by the
intraperitoneal route, a volume of 0.5 ml is administered. Centrifugation of the antivenom–
venom mixtures is not recommended because residual venom toxicity may remain in the
immunoprecipitates. After injection, deaths are recorded at 24 hours (intravenous test) or at 48
hours (intraperitoneal test) and the results analysed using Probit analysis (102), SpearmanKarber (8) or alternative procedures (such as non-parametric methods). The median effective
dose (ED50) of an antivenom is defined as the volume of antivenom that protects 50% of the
mice injected.
The ED50 can be expressed in various ways:
− mg of venom neutralized by ml of antivenom;
− µl antivenom required to neutralize the “challenge dose” of venom used;
− µl of antivenom required to neutralize one mg of venom; and
− number of LD50 of venom neutralized per ml of antivenom.
Every production laboratory and every national regulatory agency should establish the accepted
levels of neutralizing potency for the various antivenoms being produced and distributed. In this
regard, it is important to guarantee that a standardized assay is used by the manufacturing
laboratories. Since the methodology to estimate antivenom potency (i.e. ED50) varies between
laboratories and countries, manufacturers should disclose the conditions under which the potency
of their antivenoms is estimated to the corresponding regulatory agencies in the course of their
licensing and control procedures.
1
10 mice may be needed for some venoms.
69
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
The protocols for the selection and quality control of the venoms used for these potency assays
should be established in each quality control laboratory (see section 8). Venoms used in this test
should correspond to a representative pool of well-identified snake specimens collected from
various regions within the geographical range of distribution of the species in a country. These
national reference venom pools must be evaluated periodically to ensure that they have not
deteriorated (see section 8 on quality control of venoms).
Until in vitro or alternative tests of lesser severity become accepted, these venom LD50 and
antivenom ED50 assays should be performed by all manufacturers before an antivenom can be
used in humans. The assays should be conducted under conditions causing the minimal possible
suffering to the experimental animals.
15.1.5 Osmolality
Osmolality can be measured to determine the tonicity of the antivenom solution. It is
recommended that it be at least 240 mosmol/kg. Determination of osmolality is also an indirect
means to determine the quantity of salts or excipients added for formulating the batch.
15.1.6 Identification
When several types of antivenoms are produced by a single laboratory, the identity of each batch
of antivenom should be checked. Identity tests may include biological assays as well as
physicochemical and immunological tests. Double immunodiffusion assays, confronting the
antivenom with the venoms against which the antivenom is designed to act, are often used. In the
case of laboratories that use various animal species to raise antivenoms, i.e. horses and sheep, an
immunological identity test should be used to identify the mammalian species in which the
antivenoms are produced. The potency assay against venoms is another way to identify
antivenoms.
15.1.7 Protein concentration
The total protein concentration of antivenoms is performed using the Kjeldahl method for
nitrogen determination. Alternatively, several colorimetric procedures can be used, as well as
measuring absorbance at 280 nm. The presence of preservatives should be taken into account
since they may interfere with some protein determination methods (105).
The total concentration of proteins in antivenoms should preferably not exceed 10 g/dl, unless a
higher protein content is justified and authorized by the competent authority.
15.1.8 Purity
The purity of the active substance, i.e. intact immunoglobulin or immunoglobulin fragments,
should be assessed. They should constitute the great majority of the preparation, ideally greater
than 90%.
Electrophoretic methods in polyacrylamide gels (SDS–PAGE run under reducing or nonreducing conditions) are suitable for this purpose, since these techniques allow the detection and
monitoring of IgG, F(ab')2, Fab, non-immunoglobulin plasma protein contaminants (in particular
albumin), and degradation products. The electrophoretic pattern should be compared to that of a
reference preparation. A semi-quantification can be performed by calibration of the procedure.
Of particular relevance is the assessment of the albumin content which ideally should not exceed
1% of total protein content. The following approach can serve as a guide in assessing the purity
of antivenoms:
70
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
− SDS–PAGE under non-reducing conditions. This analysis provides qualitative (or, at
best, semiquantitative) information on the amounts of intact immunoglobulins, digestion
products and, importantly, on the presence of high-molecular-mass oligomers (soluble
aggregates) and low-molecular-mass contaminants (which are expected in the case of
enzymatically-digested antivenoms).
− SDS–PAGE under reducing conditions. Analysis under these conditions can provide
information on the amount of immunoglobulins and their fragments by direct
visualization of intact and/or digested immunoglobulin heavy chains.
15.1.9 Molecular-size distribution
The presence of aggregates and other components in antivenoms can be assessed by sizeexclusion liquid chromatography (gel filtration) in HPLC systems.
Densitometric analyses of chromatographic profiles allow the quantification of protein
aggregates and of the relative abundances of: intact immunoglobulins, divalent immunoglobulin
fragments (F(ab')2), monovalent immunoglobulin fragments (Fab) and dimers, as well as lowmolecular-mass enzymatic digestion products.
In intact immunoglobulin-based antivenoms this method allows quantitation of albumin as its
molecular mass (~ 66 kDa) can be resolved from the ~ 160 kDa peak of intact immunoglobulins.
15.1.10 Test for pyrogens
Antivenoms should comply with the rabbit pyrogen test where required by the local regulations.
This test is based on intravenous injection of antivenoms in the ear vein of rabbits (usually 1.0
ml per kg body mass), followed by the measurement of rectal temperature at various time
intervals after injection. The detailed procedures are described in various pharmacopoeias.
Bacterial lipopolysaccharides can also be detected by the Limulus amoebocyte lysate (LAL) test.
The test should be validated for each type of antivenom, since there have been reports of falsepositive and false-negative reactions when testing antivenoms and other plasma-derived
products. The sensitivity of this LAL test should be correlated with the rabbit pyrogen test, and
the endotoxin limits established. When regulation allows, a validated LAL test is used in place of
the rabbit pyrogen test.
15.1.11 Test for abnormal toxicity test
The abnormal toxicity test may be performed at the stage of product development but is
increasingly being abandoned in most regulations as it provides limited information for routine
quality assessment of a product. Correct implementation of GMP should provide evidence that
the product would comply with the test for abnormal toxicity.
15.1.12 Test for sterility
Antivenoms should be free of bacteria and fungi, i.e. they should be sterile. The sterility test is
performed following methodologies specified in various pharmacopoeias such as the European
pharmacopoeia.
Since antivenoms may contain preservatives in their formulation, it is necessary to “neutralize”
the preservatives before the samples are added to culture media. This is usually performed by
filtering a volume of antivenom through a 0.45-µm pore membrane, and then filtering through
the same membrane a solution that neutralizes the bacteriostatic and fungistatic effects of the
preservatives used in antivenom. The membrane is then aseptically removed and cut into two
halves. One half is added to trypticase soy broth and the other is added to thioglycolate medium.
71
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Control culture flasks are included for each medium. Flasks are incubated at 20–25 ºC (trypticase
soy broth) or at 30–35 ºC (thioglycolate) for 14 days. Culture flasks are examined daily for
bacterial or fungal growth. The number of vials tested per batch should be in compliance with
local regulations.
15.1.13 Concentration of sodium chloride and other excipients
The concentration of the various excipients or stabilizers added for formulation should be
determined using appropriate chemical methods.
15.1.14 Determination of pH
The pH of antivenom should be determined using a potentiometer.
15.1.15 Concentration of preservatives
When used in the formulation of antivenoms, the concentration of preservatives (phenol or
cresols) should be quantified. The acceptable range of preservative concentration in antivenoms
should be established and validated in each quality control laboratory. Phenol concentration
should not exceed 2.5 g/l and cresols 3.5 g/l.
Phenol concentration can be determined spectrophotometrically on the basis of the reactivity of
phenol with 4-aminoantipyrine, under alkaline conditions (pH 9.0-9.2) in the presence of
potassium ferrocyanide as oxidant. Other methods are also available. Cresols can be determined
by HPLC methods.
15.1.16 Chemical agents used in plasma fractionation
The chemical reagents used in the precipitation and purification of antivenoms, such as
ammonium sulfate, caprylic acid and others, should be removed from the final product during
diafiltration or dialysis. Limits should be established and their residual amount quantified in the
final product. Likewise, the elimination of pepsin or papain from the final preparations should be
guaranteed, especially for preparations that are maintained in liquid form, to avoid proteolytic
activity that may damage the antivenoms.
The determination of the residual amount of agents used in plasma fractionation could be
excluded from routine release testing if the process of manufacturing has been validated to
eliminate these reagents. The detection of residual reagents can also be performed on the final
bulk rather than in the final product.
15.1.17 Residual moisture (freeze-dried preparations)
Residual moisture content can be determined by several methodologies, such as:
− a gravimetric method assessing the loss of weight on heating;
− the Karl-Fischer titration, based on the principle that iodine, together with pyridine,
sulfur dioxide and methanol from the reagent react quantitatively with water; and
− thermogravimetric methods.
The methodology most commonly recommended is the Karl-Fischer titration. Every
manufacturing and quality control laboratory must establish the accepted maximum residual
moisture for their antivenom ensuring the stability of the product over its claimed shelf-life. A
residual moisture content of less than 3% is usually recommended for most freeze-dried
therapeutic biological products.
72
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
15.2 Antivenom reference preparations
The use of in-house reference preparations of antivenoms, instead of international standards, is
recommended, since the potency and specificity can only be compared with antivenoms of
similar specificity and neutralizing profile. An in-house reference preparation should be obtained
from a suitable batch of the product that has been fully evaluated by the quality control
laboratory..
15.3 Main recommendations
•
Quality control of antivenom preparations, both for product intermediates and final
product, as part of the batch release, should be performed by the manufacturers.
National regulatory agencies will review the tests performed by the manufacturer
and select which tests to develop, when required, on a case-by-case basis.
•
Quality control tests to be performed by manufacturers as part of the batch release
include: neutralization potency test against the most relevant venoms, identification,
protein concentration, purity of the active substance, content of protein aggregates
and non-IgG contaminants, pyrogen test, sterility test, concentration of excipients,
osmolality, pH, concentration of preservatives, determination of traces of agents
used in plasma fractionation, visual inspection, and, for freeze-dried preparations,
residual moisture and solubility.
•
Antivenom reference preparations reflecting specific characteristics of antivenoms
produced should be prepared by each manufacturer to be used as standards in their
laboratory settings, in particular to measure neutralization capacity of their specific
antivenom products against targeted venoms. When possible, a national reference
antivenom should be established.
16 STABILITY, STORAGE AND DISTRIBUTION OF ANTIVENOMS
16.1 Stability
Stability studies should be performed to determine the stability of antivenoms. These studies
should be done when a new product, a process change, or a new formulation is developed. They
are essential to define the shelf-life of the product and are intended to prove that the antivenom
remains stable and efficacious until the expiry date.
It has long been considered, somewhat empirically, that liquid preparations have a shelf-life of
up to 3 years at 2-8 °C, and freeze-dried preparations up to 5 years, when kept in the dark at
room temperature. Nevertheless, the actual stability of each antivenom formulation should be
appropriately determined by each manufacturer. It is highly recommended that manufacturers
perform stability studies to evaluate the possibility that their preparations could be stored for a
long period under non-refrigeration (for instance at 30 °C).
Real-time stability tests should be performed under the expected storage conditions of the
antivenom. In addition, these tests could be performed under worst-case storage conditions.
Quality control parameters are determined at regular pre-established time intervals. Essential
parameters include venom neutralization potency, turbidity and content of aggregates, among
others, since these are especially prone to alter upon storage.
Accelerated stability studies may be performed to provide early useful information on the
product stability profile, but are not a substitute for real-time data. The antivenom is exposed to
73
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
harsher conditions than usual, such as a higher temperature, and the stability is assessed over a
shorter timespan.
16.2 Storage
Antivenoms should be stored at a temperature within the range that assures stability, as found by
stability tests. This is particularly critical for liquid formulations, which usually require storage at
between 2 and 8 °C. Therefore, deviations from this temperature range, due to interruptions in
the cold chain during transportation or storage, are likely to result in product deterioration. The
design of adequate cold chain programmes, as part of the public health systems in every country,
is critical, and national protocols should be developed. The distribution policies for national
vaccination programmes can be adopted for the transportation and storage of antivenoms. The
stability of liquid preparations at temperatures higher than 2-8 °C should be evaluated and, if
needed, new formulations allowing such storage conditions should be developed.
16.3 Distribution
Adequate distribution of antivenoms is a matter of great concern in many regions of the world.
Since most of the antivenoms available are liquid preparations, the maintenance of an adequate
cold chain must be guaranteed, despite the difficulties to be encountered in rural areas of some
developing countries. National and regional health authorities should develop distribution
strategies to ensure that antivenoms are allocated to the areas where they are needed or use the
distribution channels in place for other national primary health care programmes. Both the
specificity of the antivenom and the number of vials or ampoules to be distributed should be
taken into consideration. This is particularly relevant in countries that use monospecific
antivenoms, since distribution of these products should be guided by the known distribution of
the species. To ensure an appropriate supply for clinical use, inventories should be in excess of
the estimated number of cases, to allow for unpredictable surges in local demand, accepting that
some antivenoms will not have been used by the time of their expiry date.
16.4 Main recommendations
74
•
The quality control of each antivenom batch prepared by a manufacturer should
include the potency test for neutralization of lethality (ED50).
•
In general, liquid preparations require a cold chain, whereas freeze-dried
preparations do not. However, storage conditions are product- or formulationspecific and may vary. Manufacturers should determine the stability of each
antivenom pharmaceutical preparation by conducting real-time stability studies.
•
Manufacturers should study the stability of antivenoms at the ambient
temperatures in the areas where the product will be used.
•
The distribution of antivenoms by health authorities should rely on a proper
assessment of the epidemiology of snakebite envenomings, and on the knowledge of
the geographical distribution of the most relevant venomous species. This is
particularly important for monospecific antivenoms.
•
National regulatory authorities should ask manufacturers to provide information
obtained from the preclinical assessment of all antivenom used in their territories
against the venoms found in the region or country where the product is intended to
be used.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
17 PRECLINICAL ASSESSMENT OF ANTIVENOMS
17.1 Introduction
A fundamental and ethical requirement of all new therapeutic agents intended for human use is
that their safety and efficacy should be established, initially by preclinical in vitro and in vivo
laboratory tests and, if the results of these prove satisfactory, by clinical trials in human patients.
Information supporting the physicochemical characterization of the new antivenom, such as
protein content and level of purity of the preparation should be available before clinical studies
are initiated. The assays to be performed are described under section 15, on quality control of
antivenoms.
Preclinical testing of antivenoms should be done when:
− a new antivenom is being developed;
− an existing antivenom is to be introduced for use in a new geographical region or
country.
In both cases, preclinical studies in animal models should be a regulatory requirement enforced
by the medicines regulatory authorities as part of the licensing procedures for antivenoms.
The preclinical tests of new or existing antivenoms necessitate the use of experimental rodents.
Despite reservations over the physiological relevance of these animal models to human
envenoming and the severity of these in vivo assays (sections 17.4 and 17.5), the tests for
determining venom lethality (LD50) and antivenom neutralizing capacity (ED50) are currently the
only validated means of assessing venom toxicity and antivenom neutralizing potency by both
manufacturers and regulatory authorities worldwide.
It is important to make a distinction between “essential” and “recommended” preclinical assays.
The “essential” preclinical assays consist of the overall evaluation of toxic activity of the specific
snake venoms (LD50) and the corresponding antivenom neutralizing efficacy of the overall
venom(s) toxicity (ED50). These tests are required:
− for the routine quality control of antivenom potency;
− to test the ability of a new antivenom to neutralize the venoms from snakes from the
country or region where it is going to be introduced;
− to show neutralizing efficacy of an existing antivenom against medically relevant species
in a new geographical region or country.
In summary, before any antivenom is used therapeutically in humans in a given region or
country, it should have been preclinically assessed using the “essential” assays against the
relevant snake venoms.
Preclinical testing of antivenoms also includes a number of assays whose selection depends on
the main pathophysiological effects induced by the venom to be tested. Additional tests are
therefore strongly recommended for new antivenoms and for new applications of existing
antivenoms to determine whether they are effective in eliminating the most clinically-relevant
pathophysiological effects induced by the specific venom(s) of interest.
As an example, a new antivenom developed against Echis ocellatus envenoming should be tested
for its preclinical neutralizing potency (LD50 and ED50 tests):
− before it is released for the first time for human trials; and
− for the routine quality control of the potency of subsequent batches.
75
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
It is also recommended that the first batch be preclinically tested for its ability to eliminate
venom-induced coagulopathy and haemorrhage – the most medically important effects of
envenoming by E. ocellatus.
17.2 Essential assay for preclinical testing of antivenoms: prevention of lethality
The methodology for estimating the median lethal dose (LD50) of venoms and the median
effective dose (ED50) of antivenoms is described in detail in the section on quality control of
antivenoms (section 15). The same methods used in the routine quality control of antivenoms
should be used in the preclinical testing of all new antivenoms and all new applications of
existing antivenoms.
17.3 Additional recommended assays for preclinical testing of antivenoms
It is necessary to test whether antivenoms are effective in the neutralization of the most relevant
pathophysiological effects induced by a particular venom. These “recommended” preclinical
tests are, however, not intended for the routine quality control of antivenom batches. The
relevant methods to be used are listed below.
17.3.1 Neutralization of venom haemorrhagic activity
Many venoms, especially those of vipers, exert powerful local and systemic haemorrhagic
activity which is due primarily to venom zinc metalloproteinases. These enzymes damage the
basement membrane that surrounds the endothelial cells of capillary blood vessels resulting in
bleeding into the tissues. Bleeding into the brain and other major organs is considered to be the
major lethal effect of envenoming by many viperid species (106). The minimum haemorrhagic
dose of a venom (MHD) is defined as the amount of venom (in µg dry weight) which, when
injected intradermally, induces in mice a 10-mm haemorrhagic lesion 24 hours after injection
(107, 108).
The MHD test is carried out by preparing aliquots of 50 µl of physiological saline solution
containing a range of venom doses. Mice (18–20 g body weight; 5 mice per group) are placed
under light general anaesthesia (e.g. halothane/oxygen) and the hair surrounding the injection
site is shaved. The venom solutions (50 µl) are injected intradermally in the shaved skin. After
24 hours, mice are killed using an approved humane procedure, the area of the injected skin is
removed, and the haemorrhagic lesion in the inner side of the skin is measured using calipers in
two directions with background illumination. Care should be taken not to stretch the skin. The
mean diameter of the haemorrhagic lesion is calculated for each venom dose and the MHD
estimated by plotting mean lesion diameter against venom dose and reading off the dose
corresponding to a 10-mm diameter (107, 108).
To estimate the ability of an antivenom to neutralize venom-induced haemorrhage, a “challenge
dose” of venom is selected, which corresponds to one or more MHDs. Between one and five
MHDs have been used as the challenge dose by different laboratories. The test is carried out as
above, using 5 mice per group. Mixtures of a fixed amount of venom and various dilutions of
antivenom are prepared so that the challenge dose of venom is contained in 50 µl. Controls must
include venom solutions incubated with physiological saline solution alone. Mixtures are
incubated at 37 °C for 30 min, and aliquots of 50 µl are injected intradermally in lightly
anaesthetized mice. The diameter of haemorrhagic lesions is quantified as described above, and
the neutralizing ability of antivenom, expressed as MHD-median effective dose (ED50), is
estimated as the volume of antivenom, in microlitres, which reduces the diameter of
haemorrhagic lesions by 50% when compared with the diameter of the lesion in animals injected
with the control venom/saline mixture (108).
76
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
17.3.2 Neutralization of venom necrotizing activity
Venom-induced local dermonecrosis is a major problem in human victims of snakebite and it has
long been considered important to have an assay system to evaluate the effect of an antivenom
on this pathology. However, it should be noted that the value of antivenoms in overcoming the
cytolytic effects of venoms has not yet been established; indeed, there is considerable doubt
whether antivenom is useful in obviating such effects in human victims of snakebite. This is
because venom-induced dermonecrosis occurs quickly after a bite and there is usually a
considerable delay between the envenoming of a victim and his or her arrival in hospital for
treatment. Consequently, antivenom therapy can have little or no effect in reversing the damage
(109, 110). Animal experiments in which the antivenom was administered to the animal at
different times after the venom support this opinion (110, 111).
The minimum necrotizing dose (MND) of a venom is defined as the least amount of venom (in
µg dry weight) which, when injected intradermally into groups of five lightly anaesthetized mice
(18–20 g body weight), results in a necrotic lesion of 5 mm diameter 3 days later. The method
used is the same as that for the MHD, except that the skin is examined 3 days after the
intradermal injection of the venom (107).
To estimate the ability of an antivenom to neutralize venom-induced dermonecrosis, a challenge
dose of venom is selected, usually between one and two MNDs. The test is carried out as above,
using 5 mice per group. Mixtures of a fixed concentration of venom and various dilutions of
antivenom are prepared so that the venom challenge dose is contained in 50 µl. Controls include
venom solutions incubated with physiological saline solution alone. Mixtures are incubated at
37 °C for 30 min, and aliquots of 50 µl are injected intradermally in lightly anaesthetized mice
(112, 113). The diameter of dermonecrotic lesions is quantified 3 days after injection, as
described above, and the neutralizing ability of antivenom, expressed as MND-median effective
dose (ED50), is estimated as the volume of antivenom, in microlitres, which reduces the diameter
of necrotic lesions by 50% when compared with the diameter of the lesion in mice injected with
the control venom/saline mixture.
17.3.3 Neutralization of venom procoagulant effect
Many venoms, especially from some vipers, cause consumption of coagulation factors which
results in incoagulable blood. This, combined with the haemorrhagic nature of some of these
venoms, can result in a very poor prognosis for a severely envenomed patient. Simple in vitro
methods exist to measure this venom-induced pathophysiological effect and the ability of an
antivenom to eliminate it.
The minimum coagulant dose (MCD) of a venom is defined as the least amount of venom (in mg
dry weight per litre of test solution or µg/ml) that clots either a solution of bovine fibrinogen (2
g/l) in 60 sec at 37 oC (MCD-F) and/or a standard citrated solution of human plasma (fibrinogen
content 2.8 g/l) under the same conditions (MCD-P).
For measurement of the MCD-F, 50 µl of physiological saline with final venom concentrations
ranging from 240 to 0.5 mg/l is added to 0.2 ml of bovine fibrinogen solution at 37 oC in new
glass clotting tubes. The solutions are mixed thoroughly and the clotting time recorded. The
MCD-P is estimated by adding the same venom concentrations to 0.2 ml of the standard human
plasma solution under identical conditions and recording the clotting time. In each case, the
MCD is calculated by plotting clotting time against venom concentration and reading off the
level at the 60-second clotting time (107).
To estimate the ability of an antivenom to neutralize venom procoagulant activity, a challenge
dose of venom is selected, which corresponds to one MCD-P or one MCD-F. Mixtures of a fixed
77
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
concentration of venom and various dilutions of antivenom are prepared so that the challenge
dose of venom is contained in 50 µl. Controls include venom solutions incubated with
physiological saline solution alone. Mixtures are incubated at 37 °C for 30 min, and aliquots of
50 µl are added to 0.2 ml of plasma or fibrinogen solution, as described. The formation or
absence of clots is observed during a maximum of 30 min. The minimum volume of antivenom
which completely prevents clotting is estimated and corresponds to the MCD-effective dose.
17.3.4 Neutralization of in vivo venom defibrinogenating activity
This test is a direct measure of the in vivo defibrinogenating effect of certain venoms. To
measure the minimum venom defibrinogenating dose (MDD), a wide range of venom doses is
selected and each dose, in a volume of 0.2 ml, is injected intravenously into 4 mice (18–20 g
body weight). One hour after injection, the mice are placed under terminal general anaesthesia
and bled by cardiac puncture. The blood from each animal is placed in a new glass clotting tube,
left at room temperature for 1 hour and the presence/absence of a clot recorded. The MDD is
defined as the minimum dose of venom that produces incoagulable blood in all mice tested
within 1 hour of intravenous injection.
Antivenom neutralization of the venom component(s) responsible for in vivo defibrinogenation
is estimated by incubating a challenge dose of venom, corresponding to one MDD, with different
amounts of the antivenom. Controls should include venom solutions incubated with saline
solution instead of antivenom. Mixtures are incubated at 37 °C for 30 min before injection of 0.2
ml by the intravenous route in groups of 4 mice (18–20 g body weight). After 1 hour, mice are
bled as described above, the blood is placed in new glass clotting tubes and left undisturbed for 1
hour at room temperature, after which the presence or absence of a clot is recorded. Neutralizing
ability of antivenoms is expressed as MDD-effective dose, corresponding to the minimum
volume of antivenom in which the blood samples of all injected mice showed clot formation
(113, 114).
17.3.5 Neutralization of venom myotoxic activity
The presence of myotoxic components in a venom results in the degeneration of skeletal muscle
by breaking down muscle fibres. Damage is characterized by the disruption of plasma
membranes, local infiltration of inflammatory cells and oedema. Myotoxicity is characterized by
the appearance of myoglobin in urine and by increments in the serum levels of muscle-derived
enzymes, such as creatine kinase (CK). Myotoxic phospholipase A2 (PLA2) enzymes are found
in a wide range of snake venoms. Some of these PLA2s may be primarily myotoxic, or
neurotoxic, or both. In addition, myotoxicity may occur as a consequence of ischaemia induced
in muscle fibres by the effect of haemorrhagic venom components in the microvasculature (115).
Venom myotoxic activity is determined by injecting rats or mice with various doses of venom in
a constant volume of 50 µl (using saline solution as diluent) into the right gastrocnemius muscle.
In the case of mice, groups of 5 animals of 18–20 g body weight are used per dose. Control
animals are injected with the same volume of saline solution. Tail-snip blood samples are
collected at a specific time interval (3 hr in mice), and the CK activity of serum or plasma is
determined using commercially-available diagnostic kits (116, 117). Myotoxic activity is
expressed as the minimum myotoxic dose (MMD), defined as the amount of venom that induces
an increment in serum or plasma CK activity corresponding to four times the activity in serum or
plasma of animals injected with saline solution alone. Myotoxicity can also be assessed by
histological evaluation of muscle damage after venom injection, although this is a more
expensive and more time consuming method than the CK determination.
78
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
To estimate the ability of an antivenom to neutralize venom myotoxicity, a challenge dose of
venom is selected, which corresponds to 3 MMDs. The test is carried out as above, using 5 mice
per group. Mixtures of a fixed concentration of venom and various dilutions of antivenom are
prepared so that the challenge dose of venom is contained in 50 µl. Controls include venom
solutions incubated with physiological saline solution alone. Mixtures are incubated at 37 °C for
30 min, and aliquots of 50 µl are injected into the gastrocnemius muscle, as described above.
Blood samples are collected 3 hours after injection (in the case of mice) and serum or plasma CK
activity is quantified. The neutralizing ability of antivenom, expressed as MMD-median effective
dose (ED50) is estimated as the volume of antivenom, in microlitres, which reduces the serum or
plasma CK activity by 50% when compared to the activity of animals injected with venom
incubated with saline solution only (104).
17.3.6 Neutralization of venom neurotoxic activity
Several laboratory methods for assessing venom-induced neurotoxicity have been developed
(e.g. chick biventer cervicis nerve-muscle preparation (118, 119); mouse hemidiaphragm phrenic
nerve preparation (120–124), but they are difficult to perform, require costly equipment and
expert technological help and are unlikely to be practicable for most antivenom producers.
Mouse lethality tests are usually reliable in predicting the neutralization of neurotoxic effects of
venoms.
17.4 Development of alternative assays to replace murine lethality testing
In vivo murine assays cause considerable suffering and there have been calls for the
development of alternative assays to replace the standard LD50 and ED50 tests. The controversy
relates to the balance between the clinical benefit to humans of preclinical testing against the cost
to the experimental rodents (death, pain and distress). This issue is of considerable concern and
in vivo tests should be conducted with the minimal number of animals necessary and using
protocols designed to minimize pain and suffering. There are alternative tests (124), which
reduce the need for experimental animals, use alternative non-sentient systems or use in vitro test
systems. Unfortunately, such systems cannot currently replace the rodent toxicity tests.
Consequently, the development of alternative methods to animal testing in the preclinical
evaluation of antivenoms, should be encouraged and when live animals are absolutely necessary,
anaesthesia or analgesia should be considered and evaluated to ensure that the humane benefits
of anaesthesia or analgesia to the experimental animals do not invalidate the objectives of the
assay by altering relevant physiological processes (53). The establishment of humane end-points
to reduce suffering and limiting the duration of the assays to reduce the period of animal
suffering is also encouraged, but would also need to be carefully evaluated to ensure the validity
of the results.
17.5 Limitations of preclinical assays
It is acknowledged that the in vivo and in vitro essential and recommended preclinical tests have
physiological limitations (the venom and venom/antivenom injection protocols do not represent
the natural situation, and the physiological responses of rodents to envenoming and treatment
may differ from those of humans). Such limitations make the rodent model of human
envenoming and treatment less than ideal. Care should therefore be taken to avoid simplistic
extrapolations from this assay to the clinical situation. Nevertheless, the LD50 and ED50 tests
represent the methods most widely used for assessment of antivenom potency, and a number of
clinical trials have demonstrated that the ED50 test is useful (124, 125), but not infallible (126,
127), at predicting the efficacy of antivenoms in the clinical setting. An additional value of these
79
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
tests is the assurance that antivenoms are manufactured with an accepted, quantifiable and
uniform neutralizing potency.
17.6 Main recommendations
•
Preclinical testing of antivenoms both to determine the purification profile of the
preparation and its venom(s) neutralization capacity in animal models should be a
minimum regulatory requirement to be enforced by the medicines regulatory
agencies.
•
The estimation of the ability of an antivenom to neutralize the lethal activity of
venom(s) (LD50 and ED50) is the most relevant preclinical assessment and should be
performed for all antivenoms.
•
All new antivenoms, as well as existing antivenoms to be used in new geographical
areas, should furthermore be assessed for their ability to eliminate specific
pathologies caused by the venoms of the snakes for which the antivenom has been
designed. The selection of which preclinical recommended test(s) to perform will
depend on the predominant pathophysiological effects induced by the specific snake
venom and be appropriately adapted for each antivenom. The recommended tests
are not required for quality control assessment of subsequent batches of antivenom.
•
Preclinical testing still relies heavily on the use of laboratory rodents and involves
an unsatisfactorily high degree of suffering. The working protocols should
recommend anaesthesia and analgesia to reduce suffering, where possible. Animals
should be housed, fed and handled according to approved veterinary standards.
•
Research should be promoted for the development of both refinements of the in vivo
assay protocols to reduce pain and suffering of animals, and of in vitro alternatives
to the in vivo assays to reduce the number of animals used in preclinical testing. The
results of any modified in vivo, or new in vitro protocols, should be rigorously
compared with results from existing protocols to ensure the statistical validity of the
newly developed methods.
18 CLINICAL ASSESSMENT OF ANTIVENOMS
18.1 Introduction
Antivenoms are unusual among pharmaceutical agents in that they have been used in human
patients for more than 100 years with little attention paid to clinical trials of their efficacy and
safety. However, since the 1970s it has been clearly demonstrated that it is possible to carry out
dose-finding and randomized controlled trials in human victims of snakebite envenomings.
These studies have yielded very valuable information as in the case of clinical trials of other
therapeutic agents which are generally regarded as the essential basis for regulatory approval.
The standard pathway for clinical evaluation of new therapeutic products is:
− Phase I: healthy volunteer studies – detection of unanticipated adverse events
− Phase II: limited efficacy and safety studies, often dose-finding
− Phase III: full-scale clinical evaluation, often randomized controlled trials
− Phase IV: postmarketing surveillance
80
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
The appropriateness of this pathway for antivenoms depends upon a number of factors, including
whether an antivenom is new or has been previously used in human patients and the practicality
of undertaking such studies as well as national regulatory considerations.
The conduct of clinical studies is guided by the principles set down in the international
regulations governing good clinical practice (128–130). These principles emphasize the
responsibilities of the researcher and of the organization sponsoring the research, act to protect
participants in research and ensure that the conduct of the trial is likely to lead to reliable results.
Clinical trials should be registered with an appropriate registration body, prior to
commencement.
18.1.1 Phase I studies
Conventional clinical studies using healthy volunteers are not appropriate in the case of
antivenoms1 because of the risk of anaphylactic and other reactions (e.g. pyrogenic or serum
sickness and, rarely, hypersensitivity reactions) to volunteers. Phase I studies are primarily
designed to detect unanticipated adverse events and there is extensive experience with antivenom
treatment that allows a basic understanding of its pharmacokinetics.
18.1.2 Phase II and III studies
Phase II studies are usually conducted to optimize doses, establish safety of a product and give
an indication of efficacy. Phase III studies are normally used to establish efficacy of a product,
often in comparison with an existing product, or occasionally a placebo. Since antivenoms are so
well established in the treatment of snake bite envenoming, the use of placebo controls is
ethically acceptable only where there is genuine uncertainty about whether the benefit (degree of
clinical improvement) from the antivenom outweighs the risk (potential rate of adverse events).
18.1.3 Phase IV studies
Phase IV studies are clinical surveillance studies that occur after market authorization of the
product. In view of the difficulty in performing standard clinical trials of antivenom in some
settings, this may be the only way to study safety and efficacy of an antivenom in a large number
of patients.
18.2 Clinical studies of antivenoms
Although preclinical testing may be valuable in ensuring that antivenoms neutralize the venoms
of interest, the complex effects of venoms in humans and the need to consider venom
pharmacokinetics mean that, ultimately, the efficacy and safety of antivenoms for the treatment
of human envenoming can only be determined by well designed clinical studies. Clinical studies
of antivenoms primarily address three main issues:
− assessment of the optimal initial dose of antivenom;
− assessment of efficacy of the antivenom; and
− assessment of the safety of an antivenom, particularly the incidence and severity of early
and late reactions.
Reaction rates for similar doses of a given batch of antivenom are unlikely to vary in different
geographical locations. However, following initial preclinical testing, both efficacy and dosefinding studies may need to be repeated for a new geographical location, depending upon the
1
Immunoglobulins derived from animal plasma.
81
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
similarity of the snake species in the new place with those where the antivenom was initially
tested. If the species are similar, preclinical testing indicates good neutralization, and evidence of
clinical efficacy exists in other places, postmarketing surveillance studies may be adequate.
18.2.1 Dose-finding studies
Dose-finding studies seek to establish the optimum initial dose of an antivenom required to
control envenoming. The therapeutic dose of an antivenom administered by intravenous route
depends on:
− the quantity of venom injected;
− the neutralizing potency of the antivenom; and
− the dose regimen.
The dose is calculated to neutralize a certain amount of venom and does not vary between adults
and children. Preclinical testing may be used to estimate starting doses and these dose regimens
may be evaluated in a number of ways using standard efficacy and safety end-points. Dose
regimens can be assessed using prospective observational studies (79).
In these, the proportion of patients with good clinical outcomes (for example, restoration of
blood coagulability) can be observed with different or escalating doses of antivenom.
As part of the design of the study, it is important to determine the minimum number of patients
required to establish meaningful results by using sample size calculations (131). Results may
sometimes be compared to those of previous studies (historical controls) to determine how the
efficacy or safety of a newly introduced antivenom compares with previously used antivenoms
(132). Subsequently, the minimum dose that appears to be effective can be evaluated in larger
phase II trials or compared to another antivenom or a different dose in phase III randomized
controlled trials.
18.2.2 Randomized controlled trials
Definitive phase III randomized controlled trials may require large numbers of patients because
of considerable individual variation in the clinical manifestation of envenoming. The new
antivenom is compared with the existing standard antivenom treatment or, if none exists, two
different doses of the test antivenom may be compared. Placebo controls are rarely justified
unless there is genuine uncertainty about the risk and benefits of antivenom treatment. In this
situation, as a safeguard against unnecessary morbidity in either treatment group, a restricted
sequential plan might be incorporated (133) which allows evaluation of results as the trial
progresses, as in the early trials of therapeutic tetanus antitoxin (134).
To avoid bias, patients should be randomly allocated to the groups and the study should be
blinded, at a minimum to those research personnel who are assessing the clinical response and
ideally to both investigators and participants. There should be a calculation of the number of
patients required in each trial arm to give the study sufficient statistical power. These power
calculations are based on the expected difference in outcome between the treatment groups (if
designed to demonstrate superiority of one treatment over another) or predefined limits of the
acceptable performance compared to an existing product (if designed to demonstrate that the new
antivenom is not worse than existing products (non-inferiority)).
82
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
18.2.3 Efficacy end-points for antivenom trials
The assessment criteria (end-points) used for antivenom studies should be predefined and
objective. They may be clinical or assessed by laboratory investigations. Common end-points
include mortality, time taken to restore blood coagulability (assessed by the 20-minute whole
blood clotting test) (135), other laboratory parameters such as the prothrombin time, halting of
bleeding or clinical improvement in neurotoxicity. Surrogate markers such as platelet count are
less suitable as they may be affected by complement activation resulting from antivenom
treatment itself. Patients should be observed carefully for long enough to reveal evidence of
recurrent envenoming (seen particularly with short half-life Fab antivenoms) (136).
18.2.4 Safety end-points for antivenom trials
Because antivenoms consist of foreign proteins, adverse effects are an inevitable risk in therapy.
Appropriate manufacturing steps can reduce the rate of adverse reactions. Rates of reaction are
correlated with the purity of the antivenom product and the amount of protein infused.
Continuous clinical observation at the bedside is necessary for several hours after treatment to
detect acute reactions; late adverse reactions may occur several weeks later. Accurate reaction
rates can only be assessed prospectively. Reaction rates may differ considerably between
different antivenoms, but only a small proportion are life-threatening. Studies should aim to
detect both early adverse events occurring at the time of, or within 24 hours of, antivenom
administration (such as urticaria itching, fever, hypotension or bronchospasm) and late reactions
such as serum sickness occurring between 5 and 24 days of antivenom administration (e.g. fever,
urticaria, arthralgia, lymphadenopathy, proteinuria, or neuropathy).
18.2.5 Challenges in clinical testing of antivenoms
Several particular features of snakebite make clinical testing of antivenoms challenging. These
features include the large variation in the consequences of envenoming between individuals
making it necessary to study large number of patients, difficulties in identification of the species
responsible for envenoming and the inaccessibility of areas where snakebite is sufficiently
common to provide sufficient numbers of patients to study. Clinical studies may also be
expensive, particularly if they need to be multicentre with the attendant additional complexity
and logistics. However, despite these difficulties, a number of randomized controlled trials have
been undertaken and published since 1974 (65, 78, 135, 137–142).
18.3 Post-marketing surveillance
Phase IV studies may be of much greater importance for antivenoms than is the case for other
products. A period of active post-licensing surveillance should follow:
− the introduction of a new antivenom (often a regulatory requirement);
− the introduction of an established antivenom into a new geographical area.
Although phase IV studies traditionally focus on safety, it is critical that postmarketing studies of
antivenoms examine efficacy as well as the frequency of immediate or delayed side-effects. The
combination of preclinical testing and postmarketing surveillance studies is a minimum
acceptable clinical evaluation when an existing antivenom is used in a new region.
83
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
18.3.1 Possible approaches
Passive surveillance is currently practised by some antivenom manufacturers. However,
approaches that rely upon voluntary return of questionnaires about safety and efficacy are
unlikely to provide the high quality data that are necessary. There are two potential approaches
to obtaining such data.
18.3.1.1 National or regional system for post-marketing surveillance
Countries using antivenoms should establish a national or regional system for the postmarketing
surveillance of antivenoms. Clinicians and health workers (such as those working in poison
centres) should be encouraged to report actively to national control authorities and manufacturers
any unexpected lack of clinical efficacy and adverse reactions. These should include both early
adverse events, occurring at the time of, or within 24 hours of, antivenom administration, and,
late reactions between 5 and 24 days. The mechanism for reporting (such as the use of
standardized forms), the receiving body (e.g. the national control authority), the deadline for
reporting, and the type of adverse events reportable need to be clearly defined by the authority
and will depend on its structure and resources. The manufacturer of the antivenom and the
authorities should assess these reports and, in consultation with one another and with specialists
in the field, attempt to evaluate their significance. This assessment may require the testing of
products already released and the inspection of production and control facilities and local
distribution channels. If an imported product is associated with adverse reactions, the
manufacturer and the national control authorities both in the country of distribution and from the
country of origin should be notified.
18.3.1.2 Observational studies
In certain situations, for example, the first use of an established antivenom in a new geographical
area or when routine surveillance has identified safety or efficacy concerns, there is a rationale
for setting up observational studies to ensure adequate efficacy and safety. In the case of first use
of an established antivenom in a new geographical area, such studies should follow preclinical
testing that ensures neutralization of locally important venoms. Observational studies should
carefully document the clinical responses to antivenom, the clinical outcomes and the frequency
of reactions in a cohort of patients (143).
18.3.1.3 Sentinel sites
In some settings, where postmarketing surveillance of the whole of a country may be
problematic, the use of sentinel sites may allow focusing of limited resources to maximize
surveillance effectiveness.
18.3.2 Responses to results of post-marketing studies
High quality postmarketing studies will allow clinicians, public health officials and
manufacturers to identify antivenoms with poor effectiveness, instances of incorrect use and
dosage of antivenoms and serious safety issues arising from the use of antivenoms. In some
situations, these issues may be addressed by improving training of staff in the management of
snakebite, but these studies may also allow identification of the use of an inappropriate
antivenom (144).
84
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
18.4 Main recommendations
•
Preclinical and clinical testing of antivenoms has been largely neglected in the past.
Despite challenges, clinical trials of antivenoms in human patients have proved
feasible and useful. As far as possible, trials should adhere to the principles of WHO
and International Conference on Harmonisation (ICH) good clinical practice and
should measure robust end-points.
•
National regulatory bodies should expect producers either to provide data
confirming the clinical efficacy and safety of their antivenoms, against envenoming
by local species of venomous snakes or, to support in-country clinical testing of these
products.
•
Prospective observational studies are fundamental to ensuring the efficacy and
safety of an antivenom when first used in a new geographical region.
•
Postmarketing surveillance studies should play a major role in the evaluation of
efficacy and safety of antivenoms.
19 ROLE OF NATIONAL REGULATORY AUTHORITIES
The WHO Guidelines for national regulatory authorities on quality assurance of biological
products (145, 146) state that national regulatory authorities should ensure that biological
products distributed in their territories, whether imported or manufactured locally, are of good
quality, safe and efficacious, and that manufacturers adhere to approved standards regarding
quality assurance and good manufacturing practices. The responsibilities include the
enforcement and implementation of effective national regulations, and the setting of appropriate
standards and control measures.
National regulatory authorities should increasingly play a pivotal role in ensuring the quality,
safety and efficacy of antivenoms. In the procedure for granting the marketing authorization for
an antivenom, information on the starting material, hyperimmune animal derived plasma, the
production processes and the test methods to characterize batches of the product need to be
documented as part of the dossier. An example of a summary protocol of manufacturing and
control of snake antivenom immunoglobulins to assist national regulatory authorities in
reviewing the quality of antivenom batches is shown in Appendix 2.
Assurance of the quality, safety and efficacy of snake antivenoms involves the evaluation of
information with regard to:
− the preparation of snake venom batches representative of the poisonous animals and
geographical region where the antivenom will be distributed
− the control and traceability of immunized animals and animal immunization process
− the collection, storage and transport of the hyperimmune plasma
− the fractionation of the plasma and downstream processes to produce the antivenoms;
− the test methods used to control batches of the product;
− the preclinical data supporting the expected efficacy of the products for treatment of
local envenomings;
− the clinical efficacy of locally manufactured or imported antivenoms against the species
of snakes found in the country, through active marketing surveillance.
85
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
19.1 Impact of good manufacturing practices
Implementing the principles of GMP applied to the production of therapeutic products is
acknowledged as essential for assuring the quality and safety of biological medicinal products.
This approach becomes even more important and more complex due to the nature of the
production process and the complexity and local specificities of snake envenomings. The
implementation of an appropriate quality assurance system at all stages of manufacture, should
be a pivotal element in ensuring the quality and safety of antivenoms. The following benefits
derived from the compliance with GMP:
− ensures the application of quality assurance principles at all steps involved in the
production of animal plasma and the fractionation process of antivenoms;
− reduces errors and technical problems at all stages of manufacture of plasma for
fractionation and antivenoms;
− ensures that only products which comply with quality and safety requirements, and their
marketing authorization, are released for supply;
− ensures adequate documentation and full traceability of plasma for fractionation and
antivenom production;
− enables continuous improvement in production of plasma for fractionation and
antivenoms;
− provides the basis for the national regulatory authorities to assess the compliance status
of a manufacturer of antivenoms, either local or abroad;
19.2 Establishment licence and Inspections
The enforcement and implementation of inspection and licensing regulatory systems are
fundamental tools to ensure the quality of antivenom immunoglobulins to treat snakebite
envenomings. In many countries national regulatory authorities have implemented a control
system based on licensing the establishments, inspecting them regularly, and enforcing the
implementation of the legal requirements and applicable GMP standards. This should apply to
the production of animal hyperimmune plasma for fractionation, and the manufacturing
processes of the antivenoms. The inspections and control measures should be carried out by
officials, representing the competent national regulatory authority. It is the responsibility of the
inspector from the national regulatory authority to ensure that manufacturers adhere to the
approved standards of GMP and quality assurance.
Establishments involved in all or some stages of the manufacture of antivenoms should have an
establishment licence and be inspected by the competent national regulatory authority. To obtain
the licence, the establishments need to show that their operation ensures compliance with a
defined set of requirements supporting the safety, quality and efficacy of the antivenoms. A
system control for the venoms and for the animals should be in place as part of the procedures
established for the production of animal plasma for fractionation.
Inspections may follow common inspection procedures, including an opening meeting,
inspection of main areas and activities for compliance with GMP requirements, a closing
meeting, preparation of an inspection report and follow up of any deficiencies noted. The GMP
requirements that should be covered during an inspection include verifying that all
manufacturing processes and quality control tests are clearly defined and if necessary validated;
all necessary resources are provided, including appropriately qualified and trained personnel,
adequate premises, suitable equipment and services, appropriate materials, containers and labels,
and suitable storage and transport; instructions and procedures are documented, approved,
86
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
implemented and maintained; records are kept and there is a system for handling complaints and
product recall.
A thorough inspection includes the observation of staff during performance of operations and
comparison with established standard operating procedures. The inspection should not only be
considered as checking compliance with procedures, but also as an indirect product quality
assessment by checking product-specific validation and quality control data.
A written report should summarize the main findings of the inspection including its scope, a
description of the establishment, the deficiencies listed, specified and classified (e.g. as critical,
major or minor), and a conclusion. The written report is sent to the manufacturer. The
manufacturers are requested to notify the national regulatory authority about the specific steps
which are being taken or are planned to correct the failures and to prevent their recurrence. If
necessary, follow-up inspections should be performed to verify the successful implementation of
specific corrective actions.
The national regulatory authority should have the authority to withdraw an establishment licence
if an inspection reveals critical non-compliance with the requirements or product specifications.
87
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
AUTHORS AND ACKNOWLEDGEMENTS
These Guidelines were developed under the coordination of Dr A. Padilla, Scientist, Quality
Assurance and Safety: Blood Products and Related Biologicals, World Health Organization,
Geneva.
The development of this evidence-based document has been achieved thanks to the collaboration
of a large number of experts supporting specific areas of the Guidelines. The experts who
contributed to the drafting of the document are listed below:
The first draft was prepared by the late Dr Cassian Bon, Museum National d'Histoire Naturelle,
France; Dr Thierry Burnouf, Consultant, World Health Organization; Dr Jose-Maria Gutierrez,
Instituto Clodomiro Picado, Costa Rica; Dr A. Padilla, Quality Assurance and Safety of Blood
Products and Related Biologicals, World Health Organization, Switzerland; Professor Kavi
Ratanabanangkoon, Chulabhorn Research Institute, Thailand; and Professor DA Warrell,
University of Oxford, England.
The draft was submitted for preliminary discussion to the Expert Committee on Biological
Standardization (58th Meeting). This was followed by a detailed discussion within the World
Health Organization Blood Regulators Nerwork (BRN), the members of which were:
Dr J. Epstein, Food and Drug Administration (FDA), USA; Dr A. Farrugia, Therapeutic Goods
Administration (TGA), Australia; Dr P. Ganz, Health Canada, Canada; Dr E. Griffiths, Health
Canada, Canada; Dr M. Heiden, Paul-Ehrlich Institute (PEI), Germany; Dr I. Sainte Marie,
Agence française de Sécurité sanitaire des Produits de Santé (Afssaps), France; Dr C. Schärer,
Swissmedic, Switzerland; Dr R. Seitz, Paul-Ehrlich Institute (PEI), Germany (Chairman);
Dr P. Zorzi, Agence française de Sécurité sanitaire des Produits de Santé (Afssaps), France.
Because of the importance of these Guidelines in improving production and control of
antivenoms worldwide, two World Health Organization Bi-Regional Workshops were organized
in Asia and Africa, these being the Regions where snakebite envenomings constitute a major
public health problem. The Workshops aimed to discuss the draft Guidelines with clinical
toxinologists, manufacturers, national poison centres and regulators directly involved in
manufacture and regulation of antivenoms and in the treatment of snakebite envenomings in
those Regions. The Workshops were conducted by the World Health Organization with the
collaboration of the following facilitators:
Dr T. Burnouf, Human Protein Process Sciences, France; Dr J.P. Chippaux, Institut de
Recherche pour le Développement (IRD), Bolivia; Dr J.M. Gutierrez, Instituto Clodomiro
Picado, University of Costa Rica, Costa Rica; Dr G. Müller, University of Stellenbosch, South
Africa (retired); Dr A. Padilla, World Health Organization, Switzerland; Professor H.J. de Silva,
Faculty of Medicine, University of Kelaniya, Sri Lanka; Professor P. Gopalakrishnakone,
Yong Loo Lin School of Medicine, National University of Singapore, Singapore; Dr R. Harrison,
Liverpool School of Tropical Medicine, England; Dr D. Lalloo, Liverpool School of Tropical
Medicine, England; Prof. K. Ratanabanangkoon, Chulabhorn Research Institute, Thailand;
Professor D. Theakston, Liverpool School of Tropical Medicine, England (retired);
Mr D. Williams, Australian Venom Research Unit/Nossal Institute for Global Health, School of
Medicine, University of Melbourne, Australia.
Acknowledgements are due to the following participants of the World Health Organization
Bi-Regional South-East Asian and Western Pacific Workshop on Production, Control and
Regulation of Antivenoms in Jakarta, Indonesia, for their professional contributions and useful
discussions:
88
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Mrs A. Abas, Centre for Product Registration, National Pharmaceutical Control Bureau, Ministry
of Health, Malaysia; Mrs S.S. Akter, Institute of Public Health, Bangladesh; Professor R. Alam,
Dhaka Medical College, Bangladesh; Dr S. Amudhavalli, King Institute of Preventive Medicine,
India; Mr A. Azhari, Bio Farma, Indonesia; Mr I. Baru, Drug Registration, Department of
Health, Papua New Guinea; Dr B. Bissumbhar, World Health Organization, Switzerland;
Dr Chhuo Meng, Poisons Centre, Calmette Hospital, Cambodia; Mr M. C. Dancel, Research
Institute for Tropical Medicine, Philippines; Mr A. Fernandes, Bharat Serum and Vaccines Ltd,
India; Dr S. Gnanaiah, King Institute of Preventive Medicine, India; Dr A. Gnanathasan,
Department of Clinical Medicine, Faculty of Medicine, Sri Lanka; Ms L. St. Halimah, Bio
Farma, Indonesia; Dr Heng Bun Kiet, Department of Drugs and Food, Ministry of Health,
Cambodia; Dr T.N. Huu, Pasteur Institute, Viet Nam; Mrs W. Jariyapan, Department of Medical
Sciences, Ministry of Public Health, Thailand; Dr Jing Zhang, Lanzhou Institute of Biological
Products, People's Republic of China; Dr M.V. Khadilkar, Haffkine Biopharmaceutical
Corporation Ltd, India; Professor S. Khomvilai, Queen Saovabha Memorial Institute, Thai Red
Cross, Thailand; Dr M. Kuppusamy, VINS Bioproducts Ltd, India; Mrs D. Kusmiaty, National
Agency of Drug and Food Control, Indonesia; Dr Li Jingyu, Shanghai Serum Biological
Technology Co Ltd, People's Republic of China; Dr F. Malbas, Research Institute of Tropical
Medicine, Muntinlupa, Philippines; Brig. Gen. S. M.A. Matin, Directorate of Drug
Administration, Bangladesh; Ms M.T.S. Modina, Bureau of Food and Drugs, Philippines;
Ms A. Mohamed Ariff, National Poison Center, Penang, Malaysia; Mr A. Nair, VINS
Bioproducts Ltd, India; Mr N. T. Nguyen, Poison Control Centre, Viet Nam; Ms L. Nitisaporn,
The Government Pharmaceutical Organization, Thailand; Dr L.R. Panganiban, National Poison
Management and Control Center, University of the Philippines, Philippines; Dr V.V. Pillay,
Amrita Institute of Medical Sciences & Research, India; Mr K. Ragas, CSL Biotherapies,
Victoria, Australia; Dr Y. Sano, World Health Organization Regional Office for the Western
Pacific, the Philippines; Dr B. Santoso, World Health Organization Regional Office for the
Western Pacific, the Philippines; Dr M. Shahjahan, World Health Organization, Country Office
of Indonesia; Dr Shumin Zhang, National Institute for the Control of Pharmaceutical and
Biological Products, People's Republic of China; Dr L.S. Slamet, National Agency of Drug and
Food Control, Indonesia; Mr J. Smith, CSL Biotherapies, Victoria Australia; Dr D. Sundari,
Poison Information Center, Indonesia; Dr M. Takahashi, National Institute of Infectious Diseases,
Japan; Mr B. Thapa, Ministry of Health and Population, Nepal; Dr C.L. Thapa, Ministry of
Health and Population, Dumkauli Primary Health Care Center, Nepal; Dr M. Toriba, Japan
Snake Institute, Japan; Mrs D. Vu Bach, Drug Registration Department, Ministry of Health,
Viet Nam; Professor J. White, Women's and Children's Hospital, Australia; Dr Zhou Jing, Center
for Acute Poisoning Control, Beijing, People’s Republic of China.
Acknowledgements are also due to the participants at the World Health Organization
Bi-Regional African and Eastern Mediterranean Workshop on Production, Control and
Regulation of Antivenoms in Addis Ababa, Ethiopia for their contributions and willingness to
share expertise and information:
Dr F. Aljenoobi, Food and Drug Authority, Saudi Arabia; Dr B. Allali Kouadio,
Pasteur Institute, Côte d'Ivoire; Dr S. Ansar Ahmad, Ministry of Health, Pakistan;
Dr M. Atef Abd-Elsalam, National Antivenom & Vaccine Production Center, Saudi Arabia;
Dr S. Bah, Institut National de Recherche en Santé publique, Mali; Dr S. Bokata Masika,
University of Kinshasa, Democratic Republic of Congo; Dr C.M. Baldé, Ministry of Health,
Pasteur Institute, Guinea; Dr B. Bissumbhar, World Health Organization, Switzerland;
Dr M. Chisale, World Health Organization, Regional Office for Africa, Congo; Professor
A. Diouf, Poison Centre, Health Ministry and Prevention, Senegal; Dr N. Durfa, Federal
Ministry of Health, Nigeria; Dr B. Ed Nignpense, Poisons Information & Control Centre, Ridge
Hospital, Ghana; Dr M. El-Sayed Aly Ibrahim, National Regulatory Authority, Egypt;
89
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Dr A.G. Etoundi Mballa, Hôpital Central de Yaoundé, Cameroon; Dr N.D. Fall Diop, Ministry
of Health and Prevention, Senegal; Dr V. Gomwe, Medicines Control Authority, Zimbabwe;
Mr M. Haladou, Ministry of Health, Niger; Dr G. Habib, Organization for Biological Products
and Vaccines, Egypt; Dr C. Ilonze, National Agency for Food and Drug Administration and
Control, Nigeria; Mrs N. Khalid Nomani, National Institute of Health, Pakistan; Dr F. Kolon
Diallo, Direction Nationale de la Pharmacie et du Laboratoire, Ministry of Health, Guinea;
Dr H. Langar, World Health Organization, Regional Office for the Eastern Mediterranean, Cairo;
Mr T. Makoe, South African Vaccine Producers Ltd, South Africa; Ms M. Masanja, Food and
Drugs Authority, United Republic of Tanzania; Professor A. Massougbodji, Unit of Parasitology,
Benin; Dr A. Mhenni, Pasteur Institute, Tunisia; Dr A. Mohanna Mohamed, Egyptian
Organization for Biological Products and Vaccines, Egypt; Dr F. Moin Siyoi, Pharmacy and
Poisons Board, Ministry of Health, Kenya; Professor C. Nhachi, University of Zimbabwe
Medical School, Zimbabwe; Dr G. Noreddine, Pasteur Institute, Morocco; Mr A. Okello
Okonye, National Drug Authority, Uganda; Mr K.C. Yemoa, Direction des Pharmacies et du
Médicament, Ministère de la Santé, Benin.
The second draft of these Guidelines was prepared, taking into consideration the information and
discussions held at the abovementioned Workshops. The following experts contributed to the
preparation of this draft: Dr T. Burnouf, Human Protein Process Sciences, France; Dr J. M. Gutiérrez,
Instituto Clodomiro Picado, University of Costa Rica, Costa Rica; Professor Geoff Isbister,
Menzies School of Health Research, Australia; Dr A. Padilla, World Health Organization,
Switzerland; Professor H. J. de Silva, Faculty of Medicine, University of Kelaniya, Sri Lanka;
Professor P. Gopalakrishnakone, Yong Loo Lin School of Medicine, National University of
Singapore, Singapore; Dr Rafael Otero, Programa de Ofidismo/Escorpionismo y Grupo Toxaven,
Facultad de Medicina, Universidad de Antioquia Medellín, Colombia; Dr K. Regas, CSL
Biotherapeutics, Australia; Dr R. Harrison, Liverpool School of Tropical Medicine, England;
Dr D. Lalloo, Liverpool School of Tropical Medicine, England; Professor K. Ratanabanangkoon,
Chulabhorn Research Institute, Thailand; Professor D. Theakston, Liverpool School of Tropical
Medicine, England (retired); Professor Julian White, Women's and Children Hospital, Australia.
Chapter 5 and Appendix 1 of these Guidelines, on distribution of the venomous snakes of the
highest medical importance worldwide, provides extremely valuable and detailed information
that will assist manufacturers, regulators, public health officials, governments and
nongovernmental organizations, as well as international procurement agencies, to make informed
decisions with regard to the antivenoms to be considered within a particular region, country or
territory. This Appendix was prepared by Mr D. Williams, Australian Venom Research
Unit/Nossal Institute for Global Health, School of Medicine, University of Melbourne;
Dr M. O’Shea, Australian Venom Research Unit, School of Medicine, University of Melbourne
and Dr W. Wüster, School of Biological Sciences, University of Wales. The final Draft of the
Appendix was reviewed by Dr D. Broadley, The National Museum of Zimbabwe, Zimbabwe;
Dr J.P. Chippaux, Institut de Recherche pour le Développement (IRD), La Paz, Bolivia;
Dr B. Currie, Menzies School for Health Research, Darwin, Australia; Dr J.M. Gutiérrez,
Instituto Clodomiro Picado, University of Costa Rica, San José, Costa Rica; Dr U. Kuch,
Biodiversität und Klima, Germany; Dr S. Seifert, USA; Professor D.A. Warrell, University of
Oxford, England; Professor J. White, Women's and Children Hospital, Adelaide, Australia.
90
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
WHO Secretariat:
Dr B. Bissumbhar, Quality Assurance and Safety: Blood Products and Related Biologicals,
World Health Organization, Geneva, Switzerland; Dr M. Chisale, World Health Organization,
Regional Office for Africa; Dr H. Langar, World Health Organization, Regional Office for the
Eastern Mediterranean; Dr F. Nafo-Trafore, World Health Organization Country Office of
Ethiopia; Dr A. Padilla, Quality Assurance and Safety: Blood Products and Related Biologicals,
World Health Organization, Geneva, Switzerland; Dr Y. Sano, World Health Organization,
Regional Office for the Western Pacific; Dr B. Santoso, World Health Organization, Regional
Office for the Western Pacific; Dr M. Shahjahan, World Health Organization, Country Office of
Indonesia; Dr K. Weerasuriya, World Health Organization, Regional Office for South East Asia.
91
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
REFERENCES
1. WHO Model List of Essential Medicines, 15th ed. Geneva, World Health Organization,
2007(http://www.who.int/medicines/publications/08_ENGLISH_indexFINAL_EML15.pdf).
2. Von Behring E, Kitasato S. Über das Zustande-kommen der Diphtherie-Immunität und
der Tetanus-Immunität bei Thieren. Deutsche Medizinische Wochenschrif, 1890, 16:1113–1145.
3. Phisalix C, Bertrand G. Sur la propriété antitoxique du sang des animaux vaccinés contre
le venin de vipère. Comptes Rendus de la Société de Biologie, 1894, 46:111–113.
4. Calmette A. Contribution à l'étude du venin des serpents. Immunisation des animaux et
traitement de l'envenimation ", Annales de l'Institut Pasteur, VIII, 1894:275–291.
5. Calmette A. Sur le venin des serpents et sur l'emploi du sérum antivenimeux dans la
thérapeutique des morsures venimeuses chez l'homme et chez les animaux ", Annales de l'Institut
Pasteur XII, 1897: 214–237.
6. Pope CG. The action of proteolytic enzymes on the antitoxins and proteins in immune
sera. I. True digestion of the proteins. British Journal of Experimental Pathology, 1939a,
20:132–149.
7.
Pope CG. The action of proteolytic enzymes on the antitoxins and proteins in immmune
sera. II. Heat denaturation after partial enzyme action. British Journal of Experimental
Pathology, 1939b, 20:201–212.
8. Progress in the characterization of venoms and standardization of antivenoms. Geneva,
World Health Organization, 1981 (Offset Publication No. 58).
9. Raw I et al. Antivenins in Brazil: Preparation. In: Tu AT (ed), Handbook of Natural
Toxins, Vol 5, Reptile venoms and toxins. New York, Marcel Dekker, 1991:557–581.
10. Grandgeorge M et al. Preparation of improved F(ab’)2 antivenoms. An example: new
polyvalent European viper antivenom (equine). In: Bon C, Goyffon M (eds), Envenomings and
their Treatments. Lyon, Fondation Marcel Mérieux, 1996:161–172.
11. Gutiérrez JM, León G, Lomonte B. Pharmacokinetic-pharmacodynamic relationships of
immunoglobulin therapy for envenomation. Clinical Pharmacokinetics, 2003, 42:721–741.
12. Rabies and envenomings, a neglected public health issue. Report of a Consultative
Meeting. Geneva, World Health Organization, 2007.
http://www.who.int/bloodproducts/animal_sera/Rabies.pdf).
13. Swaroop S, Grabb B. Snakebite mortality in the world. Bulletin of the World Health
Organization, 1954, 10:35–76.
14. Chippaux J-P. Snake-bites: appraisal of the global situation. Bulletin of the World Health
Organization, 1998, 76:515–524.
15. Snow RW et al. The prevalence and morbidity of snakebite and treatment-seeking
behaviour among a rural Kenyan population. Annals of Tropical Medicine and Parasitology,
1994, 88:665–671.
92
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
16. Fox S et al. Underestimation of snakebite mortality by hospital statistics in the Monaragala
District of Sri Lanka. Transactions of the Royal Society of Tropical Medicine and Hygiene, 2006,
100:693–695.
17. Hati AK et al. Epidemiology of snake bite in the district of Burdwan, West Bengal.
Journal of the Indian Medical Association, 1992, 90:145–147.
18. Pugh RN, Theakston RD. Incidence and mortality of nakebite in savanna Nigeria..
Lancet, 1980, 2:1181–1183.
19. Sharma SK et al. Impact of snakebites and determinants of fatal outcomes in southeastern
Nepal. American Journal of Tropical Medicine and Hygiene, 2004, 71:234–238.
20. Trape JF et al. High mortality from snakebite in south-eastern Senegal. Transactions of
the Royal Society of Tropical Medicine and Hygiene, 2001, 95:420–423.
21. Kasturiratne A et al. Estimating the global burden of snakebite: A literature analysis and
modelling based on regional estimates of envenoming and deaths. PLoS Medicine, 2008, 5: e218.
22. International statistical classification of diseases and related health problems, 10th ed.
Geneva, World Health Organization, 2007 (http://www.who.int/classifications/icd/en/index.html).
23. Theakston RDG, Warrell DA. Antivenoms: a list of hyperimmune sera currently
available for the treatment of envenoming by bites and stings. Toxicon, 1991, 29:1419–1470.
24. Ismail M et al. Preparation of a novel antivenom against Atractaspis and Walterinnesia
venoms. Toxicon, 2007, 49:8–18.
25. Chanhome L et al. A pilot experiment for production of Malayan krait antivenom:
immunization of rabbits with Bungarus candidus venom. Journal of Natural Toxins, 2002,
11:353–356.
26. Joseph JK et al. First authenticated cases of life-threatening envenoming by the humpnosed pit viper (Hypnale hypnale) in India. Transactions of the Royal Society of Tropical
Medicine and Hygiene, 2007, 101:85–90.
27. Ariaratnam CA et al. Frequent and potentially fatal envenoming by hump-nosed pit
vipers (Hypnale hypnale and H. nepa) in Sri Lanka: lack of effective antivenom. Transactions of
the Royal Society of Tropical Medicine and Hygiene, 2008, 102: 1120-1126
28. Chippaux JP, Williams V, White J. Snake venom variability: Methods of study, results
and interpretation. Toxicon, 1991, 29:1279–1303.
29. Warrell DA. Geographical and intraspecies variation in the clinical manifestations of
envenoming by snakes. In: Thorpe RS, Wüster W, Malhotra A. (eds) Venomous snakes. Ecology,
evolution and snakebite. Oxford, Clarendon Press, 1997:189–203.
30. Fry BG et al. Effectiveness of snake antivenom: species and regional variation and its
clinical impact. Journal of Toxicology: Toxin Reviews, 2003, 22:23–34.
31. Raweetith R, Ratanabanangkoon K. Immunochemical and biochemical comparison of
equine monospecific and polyspecific snake antivenoms. Toxicon, 2005, 45:369–375.
93
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
32. Saravia P et al. Geographic and ontogenic variability in the venom of the neotropical
rattlesnake Crotalus durissus: pathophysiological and therapeutic implications. Revista de
Biologia Tropical, 2002, 50:337–346.
33. Faure G, Bon C. Several isoforms of crotoxin are present in individual venoms from the
South American Rattlesnake, Crotalus durissus terrificus. Toxicon, 1987, 25:229–234.
34. Creer S et al. Genetic and ecological correlates of intraspecific variation in pitviper venom
composition detected using matrix-assisted laser desorption time-of-flight mass spectrometry
(MALDI-TOF-MS) and isoelectric focusing. Journal of Molecular Evolution, 2003, 56:317–329.
35. Reichenbach-Klinke H, Elkan E. Diseases of reptiles. Neptune New Jersey, TFH
Publications, 1965.
36. Cooper JE, Jackson OF. Diseases of the reptilia, 2 Vols. London, Academic Press, 1981.
37. Frye FL. Reptile care. An atlas of diseases and treatments, 2 vols. Neptune, New Jersey,
TFH Publications, 1991.
38. Shortridge KF et al. Arbovirus infections in reptiles: immunological evidence for a high
incidence of Japanese encephalitis virus in the cobra Naja naja. Transactions of the Royal
Society of Tropical Medicine and Hygiene, 1974, 68:454–460.
39. Gans C, Gans KA, Leloup P. Various aspects of venomous snake breeding on a large
scale. Acta Zoologica et Pathologica Antverpiensia, 1984, 78:177–198.
40. Mitchell MA. Snake care and husbandry. Veterinary Clinics of North America: Exotic
Animal Practice, 2004, 7:421–446.
41. Chanhome L et al. Venomous snake husbandry in Thailand. Wilderness and
Environmental Medicine Journal, 2001, 12:17–23.
42. Gutiérrez JM, Chaves F, Bolaños R. Estudio comparativo de venenos de ejemplares
recién nacidos y adultos de Bothrops asper. Revista de Biología Tropical, 1980, 28:341–351.
43. Furtado MFD et al. Comparative study of nine Bothrops snake venoms from adult female
snakes and their offspring. Toxicon, 1991, 29:219–226.
44. Alape-Girón A et al. Snake venomics of the lancead pitviper Bothrops asper: Geographic,
individual, and ontogenetic variations. Journal of Proteome Research, 2008, 7:3556–3571.
45. Kurnik D, Haviv Y, Kochva E. A snakebite by the Burrowing Asp, Atractaspis
engaddensis. Toxicon, 1999, 37:223–227.
46. Powell RL, Sanchez EE, Pérez JC. Farming of venom: Survey of snake venom extraction
facilities worldwide. Applied Herpetology, 2006, 3:1–10.
47. Nishioka SA et al. Occupational injuries with captive lance-headed vipers (Bothrops
moojeni): experience from a snake farm in Brazil. Tropical Medicine and International Health,
2000, 5:507–510.
94
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
48. Warrell DA. Envenoming, poisoning, hypersensitivity and injuries caused by animals. In:
Warrell DA, Cox TM, Firth JD (eds) Oxford Textbook of Medicine, 4th ed. Oxford, Oxford
University Press, 2005.
49. Wüster W, McCarthy CJ. Venomous snake systematics: Implications for snakebite
treatment and toxinology. In: Envenomings and their Treatments, 1996:13–23. Bon C, Goyffon
M. (eds). France Lyon: Fondation Mérieux.
50. Angulo Y, Estrada R, Gutiérrez JM. Clinical and laboratory alterations in horses during
immunization with snake venoms for the production of polyvalent (Crotalinae) antivenom.
Toxicon, 1997, 35:81–90.
51. Landon J, Smith D. Merits of sheep antisera for antivenom manufacture. Journal of
Toxicology: Toxin Reviews, 2003, 22:15–22.
52. Landon J, Woolley JA, McLean C. Antibody production in the hen. In: Landon J, Chard
T (eds.) Therapeutic antibodies. London, Springer-Verlag, 1995:47–68.
53. Theakston RDG, Warrell DA, Griffiths E. Report of a WHO workshop on the
standardization and control of antivenoms. Toxicon, 2003, 41:541–557.
54. Guidelines on tissue infectivity distribution in transmissible spongiform encephalopathies.
Geneva, World Health Organization 2006:1–53
(http://www.who.int/bloodproducts/cs/TSEPUBLISHEDREPORT.pdf).
55. Moroz-Perlmutter C et al. Detoxification of snake venoms and venom fractions by
formaldehyde. Proceedings of the Society for Experimental Biology and Medicine, 1963,
112:595–598.
56. Freitas TV et al. Immunization of horses with Crotalus durissus terrificus (South
American rattlesnake) venom. A comparison of four different procedures. Revista brasileira de
pesquisas medicas e biologicas/Sociedade Brasileira de Biofisica, 1991, 24:281–290.
57. Pratanaphon R et al. Production of highly potent horse antivenom against the Thai cobra
( Naja kaouthia). Vaccine, 1997, 15:1523–1528.
58. Chotwiwatthanakun C et al. Production of potent polyvalent antivenom against three
elapid venoms using a low dose, low volume, multi-site immunization protocol. Toxicon, 2001,
39:1487–1494.
59. Quality assurance of pharmaceuticals. A compendium of guidelines and related materials.
Vol 2, 2nd updated ed. Good manufacturing practices and inspection. Geneva, World Health
Organization, 2007 (http://www.who.int/medicines).
60. Guidelines on viral inactivation and removal procedures intended to assure the viral safety
of human blood plasma products. Geneva, World Health Organization, 2004 (WHO Technical
Report Series No. 924): http://www.who.int/bloodproducts/publications/WHO_TRS_924_A4.pdf.
61. Recommendations for the production, control and regulation of human plasma for
fractionation. Geneva, World Health Organization, 2007, Annex 4 (WHO Technical Report
Series No. 941): http://www.who.int/bloodproducts/publications/TRS941Annex4blood.pdf.
95
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
62. Note for guidance on production and quality control of animal immunoglobulins and
immunsera for human use (CPMP/BWP/3354/99). London: European Agency for the Evaluation of
Medicinal Products (EMEA), 2002:1–14 (http://www.emea.europa.eu/pdfs/human/bwp/335499en.pdf).
63. Bolaños R, Cerdas L. Producción y control de sueros antiofídicos en Costa Rica. Boletín
de la Oficina Sanitaria Panamericana, 1980, 88:189–196.
64. Rojas G, Jiménez JM, Gutiérrez JM. Caprylic acid fractionation of hyperimmune horse
plasma: description of a simple procedure for antivenom production. Toxicon, 1994, 32:351–363.
65. Otero R et al. A randomized blinded clinical trial of two antivenoms, prepared by caprylic
acid or ammonium sulfate fractionation of IgG, in Bothrops and Porthidium snake bites in
Colombia: correlation between safety and biochemical characteristics of antivenoms. Toxicon,
1999, 37:895–908.
66. Steinbuch M, Audran R. The isolation of IgG from mammalian sera with the aid of
caprylic acid. Archives of Biochemistry and Biophysics, 1969, 134:279–284.
67. Dos Santos MC et al. Purification of F(ab’)2 anti-snake venom by caprylic acid: a fast
method for obtaining IgG fragments with large neutralization activity, purity and yield. Toxicon,
1989, 27:297–303.
68. Gutiérrez JM et al. Pan-African polyspecific antivenom produced by caprylic acid
purification of horse IgG: an alternative to the antivenom crisis in Africa. Transactions of the
Royal Society of Tropical Medicine and Hygiene, 2005, 99:468–475.
69. Otero-Patiño R et al. A randomized, blinded, comparative trial of one pepsin-digested and
two whole IgG antivenoms for Bothrops snakebites in Urabá, Colombia. American Journal of
Tropical Medicine and Hygiene, 1998, 58:183–189.
70. Jones RGA, Landon J. A protocol for ‘enhanced pepsin digestion’: a step by step method
for obtaining pure antibody fragments in high yield from serum. Journal of Immunological
Methods, 2003, 275:239–250.
71. Raweerith R, Ratanabanangkoon K. Fractionation of equine antivenom using caprylic
acid precipitation in combination with cationic ion-exchange chromatography. Journal of
Immunological Methods, 2003, 282:63–72.
72. Al-Abdulla I et al. Formulation of a liquid ovine Fab-based antivenom for the treatment of
envenomation by the Nigerian carpet viper (Echis ocellatus). Toxicon, 2003, 42:399–404.
73. Saetang T et al. Quantitative comparison on the refinement of horse antivenom by salt
fractionation and ion-exchange chromatography. Journal of Chromatography, 1997, 700:233–239.
74. Sullivan JB jr, Russell FE. Isolation and purification of antibodies to rattlesnake venom
by affinity purification. Proceedings of the Western Pharmacology Society, 1982 , 25:185–189.
75. Magos, L. Review on the toxicity of ethylmercury, including its presence as a preservative
in biological and pharmaceutical products. Journal of Applied Toxicology, 2001, 21:1–5.
96
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
76. Guidelines on regulatory expectations related to the elimination, reduction or replacement
of thiomersal in vaccines. Geneva, World Health Organization, 2004, Annex 4 (WHO Technical
Report Series, No. 926).
77. Pikal MJ. Freeze drying. In: Encyclopedia of pharmaceutical technology. New York,
Marcel Dekker, 2002:1299–1326.
78. Meyer WP et al. First clinical experiences with a new ovine Fab Echis ocellatus snakebite
antivenom in Nigeria: randomised comparative trial with Institute Pasteur serum (Ipser) Africa
antivenom. American Journal of Tropical Medicine and Hygiene, 1997, 56:291–300.
79. Ariaratnam CA et al. A new monospecific ovine Fab fragment antivenom for treatment of
envenoming by the Sri Lankan Russell’s Viper (Daboia russelii russelii): a preliminary dosefinding pharmacokinetic study. American Journal of Tropical Medicine and Hygiene, 1999,
61:259–265.
80. Ho M et al. Pharmacokinetics of three commercial antivenoms in patients envenomed by
the Malayan pit viper, Calloselasma rhodostoma, in Thailand. American Journal of Tropical
Medicine and Hygiene, 1990, 42:260–266.
81. Scherrmann JM. Antibody treatment for toxin poisoning: recent advances. Journal of
Toxicology – Clinical Toxicology, 1994, 32:363–375.
82. Audebert F et al. Viper bites in France: clinical and biological evaluation; kinetics of
envenomations. Human & Experimental Toxicology, 1994, 13:683–688.
83. Choumet V et al. New approaches in antivenom therapy. Advances in Experimental
Medicine and Biology, 1996, 391:515–520.
84. Boyer LV, Seifert SA, Cain JS. Recurrence phenomena after immunoglobulin therapy for
snake envenomations: guidelines for clinical management with crotaline Fab antivenom. Annals
of Emergency Medicine, 2001, 37(Part 2):196–201.
85. Rivière G et al. Effect of antivenom on venom pharmacokinetics in experimentally
envenomed rabbits: toward an optimization of antivenom therapy. Journal of Pharmacology and
Experimental Therapeutics, 1997, 281:1–8.
86. Rivière G et al. Absorption and elimination of viper venom after antivenom
administration. Journal of Pharmacology and Experimental Therapeutics, 1998, 285:490–495.
87. Burnouf T et al. Assessment of the viral safety of antivenoms fractionated from equine
plasma. Biologicals, 2004, 32:115–128.
88. Dürrwald R et al. Meta-analysis of putative human bornavirus sequences fails to provide
evidence implicating Borna disease virus in mental illness. Reviews in Medical Virology, 2007,
17:181–203.
89. Dichtelmuller H, Rudnick D, Kloft M. Inactivation of lipid enveloped viruses by
octanoic acid treatment of immunoglobulin solution. Biologicals, 2002, 30:135–142.
90. Korneyeva M et al. Enveloped virus inactivation by caprylate: A robust alternative to
solvent-detergent treatment in plasma derived intermediates. Biologicals, 2002, 30:153–162.
97
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
91. Parkkinen J et al. A modified caprylic acid method for manufacturing immunoglobulin G
from human plasma with high yield and efficient virus clearance. Vox Sanquinis, 2006, 90:97–104.
92. Reid KG et al. Potential contribution of mild pepsin treatment at pH4 to the viral safety of
human immunoglobulin products. Vox Sanguinis, 1988, 55:75–80.
93. Omar A et al. Virus inactivation by pepsin treatment at pH 4 of IgG solutions: factors
affecting the rate of virus inactivation. Transfusion, 1996, 36:866–872.
94. Bos OJ et al. Virus validation of pH 4-treated human immunoglobulin products produced
by the Cohn fractionation process. Biologicals, 1998, 26:267–276.
95. Horowitz B et al. Inactivation of lipid-enveloped viruses in labile blood derivatives by
unsaturated fatty acids. Vox Sang, 1988, 54:14–20.
96. Lundblad JL, Seng RL. Inactivation of lipid-enveloped viruses in proteins by caprylate.
Vox Sanguinis, 1991, 60:75–81.
97. Burnouf T et al. Assessment of viral inactivation during pH 3.3 pepsin digestion and
caprylic acid treatment of antivenoms. Biologicals, 2007, 35:329–334.
98. Mpandi M et al. Partitioning and inactivation of viruses by the caprylic acid precipitation
followed by a terminal pasteurization in the manufacturing process of horse immunoglobulins.
Biologicals, 2007, 35:335–341.
99. Lazar A et al. Inactivation of West-Nile virus during peptic cleavage of horse plasma IgG.
Biologicals, 2002, 30:163–165.
100. Cameron-Smith R et al. The removal of viruses during the purification of equine antisera
using filtration aids Hyflo Super-Cel and Fulmont Super A. Biologicals, 2000, 28:169–174.
101. Burnouf T et al. Place of nanofiltration for assuring viral safety of biologicals. Current
Nanoscience, 2005, 1:189–201.
102. Finney DJ. Probit analysis, 3rd ed. Cambridge, Cambridge University Press, 1971.
103. The biological potency assay for antivenom preparations. Expert Committee on Biological
Standardization, BS/90.1641. Geneva, World Health Organization, 1990.
104. Rojas E et al. Neutralization of four Peruvian Bothrops sp snake venoms by polyvalent
antivenoms produced in Perú and Costa Rica: preclinical assessment. Acta Tropica, 2005, 93:85–95.
105. Tuck S, Ng R. Protein assays. In: Rodroguez-Diaz R, Wehr T, Tuck S eds. Analytical
techniques for biopharmaceutical development. New York, Marcel Dekker, 2005:14–24.
106. Reid HA, Theakston RDG. The management of snakebite. Bulletin of the World Health
Organization, 1983, 61:885–895.
107. Theakston RDG, Reid HA. Development of simple standard assay procedures for the
characterisation of snake venoms. Bulletin of the World Health Organization, 1983, 61:949–956.
108. Gutiérrez JM et al. Neutralization of proteolytic and hemorrhagic activities of Costa
Rican snake venoms by a polyvalent antivenom. Toxicon, 1985, 23:887–893.
98
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
109. Reid HA. Cobra bites. British Medical Journal, 1964, ii:540–545.
110. Gutiérrez JM et al. Neutralization of local tissue damage induced by Bothrops asper
(terciopelo) snake venom. Toxicon, 1998, 36:1529–1538.
111. Iddon D, Theakston RDG, Ownby CL. A study of the pathogenesis of local skin necrosis
induced by Naja nigricollis (spitting cobra) venom using simple histological staining techniques.
Toxicon, 1987, 25:665–672.
112. Laing GD et al. BIASG (Butantan Institute Antivenom Study Group). Comparison of the
potency of three Brazilian Bothrops antivenoms using in vivo rodent and in vitro assays.
Toxicon, 1992, 30:1219–1225.
113. Theakston, RDG. Characterization of venoms and standardization of antivenoms. In:
Harris, JB (ed). Natural toxins, animal, plant and microbial. Oxford, Clarendon Press, Oxford
Science Publications, 1986:287–303.
114. Gené JA et al. Comparative study on coagulant, defibrinating, fibrinolytic and
fibrinogenolytic activities of Costa Rican crotaline snake venoms and their neutralization by a
polyvalent antivenom. Toxicon, 1989, 27:841–848.
115. Gutiérrez JM et al. Skeletal muscle necrosis and regeneration after injection of BaH1, a
hemorrhagic metalloproteinase isolated from the venom of the snake Bothrops asper
(terciopelo). Experimental and Molecular Pathology, 1995, 62:28–41.
116. Gutiérrez JM et al. Experimental myonecrosis induced by the venoms of South American
Micrurus (Coral snakes). Toxicon, 1992, 30:1299–1302.
117. Alam JM et al. Myotoxic characteristics of snake venoms; myonecrosis induced in albino
rats by crude venoms and the purified myotoxic component, phospholipase A2. Journal of
College of Physicians and Surgeons Pakistan, 2002, 12:398–404.
118. Ginsborg BL, Warriner JN. The isolated chick biventer cervicis nerve-muscle
preparation. Bristish Journal of Pharmacology, 1960, 15:410–411.
119. Harvey AL et al. Screening of snake venoms for neurotoxic and myotoxic effects using
simple in vitro preparations from rodents and chicks. Toxicon, 1994, 32:257–265.
120. Bulbring E. Observations on the isolated phrenic nerve diaphragm of the rat. British
Journal of Pharmacolology, 1946, 1:38–61.
121. Kitchen I. Neuromuscular blocking drugs and the rat phrenic nerve hemidiaphragm
preparation. In: Textbook of in Vitro Pharmacology, 3rd ed. Oxford, Blackwell Scientific,
2009:79–83.
122. Jones RGA, Lee L, Landon J. The effects of specific antibody fragments on the
‘irreversible’ neurotoxicity induced by Brown snake (Pseudonaja) venom. British Journal of
Pharmacology, 1999, 126:581–584.
123. Crachi MT, Hammer LW, Hodgson WC. A pharmacological examination of venom
from the Papuan taipan (Oxyuranus scutellatus canni). Toxicon, 1999, 37:1721–1734.
99
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
124. Sells PG. Animal experimentation in snake venom research and in vitro alternatives.
Toxicon, 2003, 42:115–133.
125. Theakston RDG et al. Treatment of snake bites by Bothrops species and Lachesis muta in
Ecuador: laboratory screening of candidate antivenoms. Transactions of the Royal Society of
Tropical Medicine and Hygiene, 1995, 89:550–554.
126. Keegan HE et al. Southeast Asian snake bite antivenin studies. Japan, 406th Medical
Laboratory, Control Department of Entomology, US Army Medical Command, 1964.
127. Warrell DA et al. Comparison of Pasteur and Behringwerke antivenoms in envenoming
by the carpet viper (Echis carinatus). British Medical Journal, 1980, 280:607–609.
128. Guidelines for good clinical practice (GCP). International Conference on Harmonisation of
Technical Requirements for Registration of Pharmaceuticals for Human Use (ICH), 1996. Step 5.
(http://www.ich.org).
129. Guidelines for good clinical practice (GCP) for trials on pharmaceutical products. In: WHO
Expert Committee on the use of Essential Drugs, Sixth report. Geneva, World Health
Organization, 1995, Annex 3 (WHO Technical Report Series, No. 850).
130. Handbook for good clinical research practices (GCP). Geneva, WHO, 2005
(http://www.who.int/medicinedocs/index/assoc/s14084e/s14084e.pdf).
131. Machin D et al. Sample size tables for clinical studies, 3rd ed. Chichester, Wiley
Blackwell, 2009.
132. Visser et al. Failure of a new antivenom to treat Echis ocellatus snake bite in rural Ghana:
the importance of quality surveillance. Transactions of the Royal Society of Tropical Medicine
and Hygiene, 2008, 102: 445–450.
133. Armitage P. Sequential medical trials, 2nd ed. New York, John Wiley, 1975.
134. Vakil BJ et al. Therapeutic trial of extracisternal human tetanus immune globulin in
clinical tetanus. Transactions of the Royal Society of Tropical Medicine and Hygiene, 1979,
73:579–583.
135. Smalligan R et al. Crotaline snake bite in the Ecuadorian Amazon: randomised double
blind comparative trial of three South American polyspecific antivenoms. British Medical
Journal, 2004, 329:1129.
136. Ariaratnam CA et al. An open, randomized comparative trial of two antivenoms for the
treatment of envenomings by Sri Lankan Russell's viper (Daboia russelli russelli). Transactions
of the Royal Society of Tropical Medicine and Hygiene, 2001, 95:74–80.
137. Warrell DA et al. Bites by the saw-scaled or carpet viper (Echis carinatus): trial of two
specific antivenoms. British Medical Journal, 1974, 4:437–440.
138. Warrell DA et al. Randomized comparative trial of three monospecific antivenoms for
bites by the Malayan pit viper (Calloselasma rhodostoma) in southern Thailand: clinical and
laboratory correlations. American Journal of Tropical Medicine and Hygiene 1986, 35:1235–1247.
100
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
139. Cardoso JL et al. Randomized comparative trial of three antivenoms in the treatment of
envenoming by lance-headed vipers (Bothrops jararaca) in Sao Paulo, Brazil. Quarterly Journal
of Medicine, 1993, 86:315–325.
140. Jorge MT et al. A randomized 'blinded' comparison of two doses of antivenom in the
treatment of Bothrops envenoming in Sao Paulo, Brazil. Transactions of the Royal Society of
Tropical Medicine and Hygiene, 1995, 89:111–114.
141. Otero R et al. A randomized double-blind clinical trial of two antivenoms in patients
bitten by Bothrops atrox in Colombia. The Regional Group on Antivenom Therapy Research
(REGATHER). Transactions of the Royal Society of Tropical Medicine and Hygiene, 1996,
90:696–700.
142. Otero R et al. Efficacy and safety of two whole IgG polyvalent antivenoms, refined by
caprylic acid fractionation with or without beta-propiolactone, in the treatment of Bothrops asper
bites in Colombia. Transactions of the Royal Society of Tropical Medicine and Hygiene, 2006,
100:1173–1182.
143. Bush et al. Crotalidae polyvalent immune Fab (ovine) antivenom is efficacious for
envenomations by Southern Pacific rattlesnakes (Crotalus helleri). Annals of Emergency
Medicine, 2002, 40:619–624.
144. Warrell DA. Unscrupulous marketing of snakebite antivenoms in Africa and Papua New
Guinea: Choosing the right product. Transactions of the Royal Society of Tropical Medicine and
Hygiene, 2008, 102:397-399
145. Guidelines for national authorities on quality assurance for biological products. In: WHO
Expert Committee on Biological Standardization. Forty-second Report. Geneva, World Health
Organization, 1992, Annex 2 (WHO Technical Report Series, No. 822).
146. Regulation and licensing of biological products in countries with newly developing
regulatory authorities. In: WHO Expert Committee on Biological Standardization Forty-fifth
Report. Geneva, World Health Organization, 1995, Annex 1 (WHO Technical Report Series, No.
858).
101
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
© World Health Organization
APPENDIX 1
Worldwide distribution of medically important venomous snakes
Venomous snakes are widely distributed especially in tropical countries, from sea level to
altitudes of up to 4 900 metres (Gloydius himalayanus). The European adder (Vipera berus)
enters the Arctic Circle, and the Argentine Yararanata (Bothrops ammodytoides) occurs to 47 ËšS
and is the most southerly occurring venomous snake. No other venomous species occur in cold
regions such as the Arctic, Antarctic and north of about latitude 51 ºN in North America
(Newfoundland, Nova Scotia).
This Appendix lists venomous snake species considered to represent the greatest threat to
public health in various countries, territories and other areas or regions around the world.
Only species that fall into one of the two categories listed below are shown, and category listings
are in alphabetical order according to taxonomic family, genus and species. The intention in
categorizing these medically important snakes into two groups is to provide users of the
Guidelines with a prioritized listing. Species listed in Category 1 within a country, territory or
area should be considered as being of highest priority for antivenom production on the basis that
available knowledge implicates them as being responsible for the greater burden in that
particular setting.
Definitions of Category 1 and Category 2 are:
CATEGORY 1: Highest medical importance
Definition: Highly venomous snakes which are common or widespread and cause
numerous snakebites, resulting in high levels of morbidity, disability or mortality.
CATEGORY 2: Secondary medical importance
Definition: Highly venomous snakes capable of causing morbidity, disability or death, for
which exact epidemiological or clinical data may be lacking; and/or which are less
frequently implicated (due to their activity cycles, behaviour, habitat preferences or
occurrence in areas remote to large human populations).
There are numerous other venomous species that rank as lesser threats in countries, territories
and other areas listed here, and interested readers should refer to herpetological references in
these guidelines. It should be noted that over time, as more information becomes available, new
species may be added to these lists, and/or some species, currently defined within Category 1 or
Category 2 may be re-ranked.
It should also be noted that the organization of countries, territories and other areas in this
Appendix does not follow the WHO regional organization, but is instead arranged biogeographically in alphabetical order of country, territory or geographical area. This approach
was necessary to reflect the geographical distribution of major groups of venomous snakes
throughout the world. For example, the venomous snakes of the eastern Indonesian Province of
Papua have bio-geographical origins in Australo-Papua, and are evolutionarily distinct from the
venomous snakes of Asian origin that occur west of Wallace’s Line which runs south of the
Philippines, between Borneo and Sulawesi, and between Bali and Lombok separates the
biogeographical regions of Asia and Australia. For this reason, we have listed the medically
important snakes of Indonesian Papua in the Australo-Papuan region, rather than the South-East
Asian region.
102
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Users of this Appendix should also recognize that the relative risk of injury from a particular
species may vary from one country, territory or area to another. For this reason, some species
that have been listed under Category 1 in one country, territory or area may have been listed
under Category 2 in another country territory or area, as a reflection of the different risk posed
by that species in different locations. Assignment to Category 1 or Category 2 was based in some
cases on the relative importance of a species as a cause of snakebite. In Europe for example, the
overall incidence of snakebite is trivial compared to that in West Africa or India, but where a
European species (such as Vipera berus) is a major (or sole) cause of envenoming where it
occurs, this warrants ranking it as a medically important species in that setting.
AFRICA AND THE MIDDLE EAST
Island populations
Off the coast of Africa, there are no medically important snakes in Mauritius, Réunion,
Rodrigues, the Comoros, the Canary Islands, the Cape Verde Islands or the Seychelles. The
islands that do have venomous snakes include the Lamu group, Zanzibar, Pemba and Mafia
Islands, the Bazaruto Archipelago and Inhaca Island, São Tomé, Principe, Bioko (Fernando Po)
and Dahlak Islands. The venomous snakes on these islands tend to be similar to those on the
adjacent mainland. A colubrid, Madagascarophis meridionalis, and perhaps other species of the
same genus, are the only terrestrial snakes of possible, if minimal, medical importance found in
Madagascar.
North Africa
Algeria:
Cat 1:
Elapidae: Naja haje; Viperidae: Cerastes cerastes; Daboia mauritanica1
Cat 2:
Viperidae: Daboia deserti1; Echis leucogaster; Macrovipera lebetina; Vipera
latastei
Egypt:
Cat 1:
Elapidae: Naja haje; Viperidae: Cerastes cerastes; Echis coloratus (east), Echis
pyramidum;
Cat 2:
Atractaspididae: Atractaspis engaddensis (Sinai); Elapidae: Naja nubiae1;
Walterinnesia aegyptia (Sinai); Viperidae: Pseudocerastes fieldi
The Libyan Arab Jamahiriya:
Cat 1:
Elapidae: Naja haje; Viperidae: Cerastes cerastes; Echis pyramidum
Cat 2:
Viperidae: Daboia deserti1
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
103
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Morocco:
Cat 1:
Elapidae: Naja haje; Viperidae: Bitis arietans; Cerastes cerastes; Daboia
mauritanica1
Cat 2:
Viperidae: Echis leucogaster; Vipera latastei
Tunisia:
Cat 1:
Viperidae: Daboia mauritanica1
Cat 2:
Elapidae: Naja haje; Viperidae: Cerastes cerastes; Daboia deserti1; Echis
leucogaster; Macrovipera lebetina; Vipera latastei
Western Sahara:
Cat 1:
Viperidae: Cerastes cerastes
Cat 2:
Elapidae: Naja haje; Viperidae: Bitis arietans
Central sub Saharan Africa
Angola:
Cat 1:
Elapidae: Dendroaspis jamesoni, Dendroaspis polylepis; Naja anchietae1, Naja
melanoleuca, Naja nigricollis; Viperidae: Bitis arietans, Bitis gabonica1
Cat 2:
Atractaspididae: Atractaspis bibronii, Atractaspis irregularis; Colubridae:
Dispholidus typus; Thelotornis capensis, Thelotornis kirtlandii (north); Elapidae:
Naja christyi1 (Cabinda), Naja mossambica (south), Naja nigricincta1 (south-west);
Pseudohaje goldii; Viperidae: Atheris squamigera; Bitis nasicornis (Cabinda)
Burundi:
Cat 1:
Elapidae: Naja nigricollis; Naja melanoleuca; Viperidae: Bitis arietans
Cat 2:
Atractaspididae: Atractaspis bibronii, Atractaspis irregularis; Colubridae:
Dispholidus typus; Thelotornis mossambicanus1; Elapidae: Dendroaspis jamesoni;
Viperidae: Bitis gabonica1, Bitis nasicornis
The Central African Republic:
Cat 1:
Elapidae: Dendroaspis jamesoni, Dendroaspis polylepis; Naja haje, Naja
nigricollis; Viperidae: Bitis arietans, Bitis gabonica1; Echis ocellatus, Echis
pyramidum
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Thelotornis
kirtlandii; Elapidae: Naja annulata1, Naja melanoleuca2; Pseudohaje goldii;
Viperidae: Atheris broadleyi, Atheris squamigera; Bitis nasicornis1
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
The medical importance of this species may be higher in the primary forest zone of the south-western Central
African Republic, and in some secondary forest mosaic zones elsewhere in the Central African Republic.
2
104
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Chad:
Cat 1:
Elapidae: Naja haje, Naja nigricollis; Viperidae: Bitis arietans (south); Echis
ocellatus (south)
Cat 2:
Colubridae: Dispholidus typus; Elapidae: Naja katiensis, Naja nubiae1; Viperidae:
Cerastes cerastes
The Congo:
Cat 1:
Elapidae: Dendroaspis jamesoni; Naja melanoleuca; Viperidae: Bitis gabonica1,
Bitis nasicornis
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Thelotornis
kirtlandii; Elapidae: Naja annulata1, Naja christyi1, Naja nigricollis; Pseudohaje
goldii; Viperidae: Atheris squamigera; Bitis arietans
The Democratic Republic of the Congo:
Cat 1:
Elapidae: Dendroaspis jamesoni; Naja melanoleuca, Naja nigricollis; Viperidae:
Bitis arietans, Bitis gabonica1, Bitis nasicornis
Cat 2:
Atractaspididae: Atractaspis bibronii, Atractaspis irregularis; Colubridae:
Dispholidus typus; Thelotornis capensis, Thelotornis kirtlandii; Elapidae:
Dendroaspis polylepis; Naja anchietae1 (Katanga pedicle), Naja annulata1, Naja
christyi1, Naja haje (north); Pseudohaje goldii; Viperidae: Atheris squamigera
Equatorial Guinea:
Cat 1:
Elapidae: Dendroaspis jamesoni; Naja melanoleuca; Viperidae: Bitis gabonica1,
Bitis nasicornis
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Thelotornis kirtlandii;
Elapidae: Naja annulata1; Pseudohaje goldii; Viperidae: Atheris squamigera
Gabon:
Cat 1:
Elapidae: Dendroaspis jamesoni; Naja melanoleuca, Naja nigricollis; Viperidae:
Bitis gabonica1, Bitis nasicornis
Cat 2:
Viperidae: Atractaspis irregularis; Colubridae: Thelotornis kirtlandii; Elapidae:
Naja annulata1; Pseudohaje goldii; Viperidae: Atheris squamigera; Bitis arietans
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
105
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Rwanda:
Cat 1:
Elapidae: Dendroaspis jamesoni; Naja nigricollis; Viperidae: Bitis arietans
Cat 2:
Atractaspididae: Atractaspis bibronii, Atractaspis irregularis; Colubridae:
Dispholidus typus; Thelotornis kirtlandii; Elapidae: Dendroaspis polylepis; Naja
annulata1, Naja melanoleuca; Pseudohaje goldii; Viperidae: Bitis gabonica1, Bitis
nasicornis
East sub Saharan Africa
Djibouti:
Cat 1:
Viperidae: Echis pyramidum
Cat 2:
Atractaspididae: Atractaspis fallax; Colubridae: Dispholidus typus; Elapidae: Naja
pallida; Viperidae: Bitis arietans
Eritrea:
Cat 1:
Elapidae: Dendroaspis polylepis; Naja haje; Viperidae: Bitis arietans; Echis
pyramidum
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Elapidae:
Naja nubiae1; Viperidae: Echis megalocephalus
Ethiopia:
Cat 1:
Elapidae: Dendroaspis polylepis; Naja ashei1 (south-east), Naja haje, Naja
nigricollis; Viperidae: Bitis arietans; Echis pyramidum
Cat 2:
Atractaspididae: Atractaspis fallax, Atractaspis irregularis (Mount Bizen);
Colubridae: Dispholidus typus; Elapidae: Naja melanoleuca, Naja pallida;
Viperidae: Bitis parviocula
Kenya:
Cat 1:
Elapidae: Dendroaspis angusticeps, Dendroaspis polylepis; Naja ashei1 (north &
east), Naja haje, Naja nigricollis; Viperidae: Bitis arietans; Echis pyramidum
Cat 2:
Atractaspididae: Atractaspis bibronii, Atractaspis fallax, Atractaspis irregularis;
Colubridae: Dispholidus typus; Thelotornis mossambicanus1, Thelotornis
usambaricus1 (east coast); Elapidae: Dendroaspis jamesoni; Naja melanoleuca
(west & coastal forest), Naja pallida (north & east); Pseudohaje goldii; Viperidae:
Atheris squamigera; Bitis nasicornis, Bitis gabonica1 (west)
1
106
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Malawi:
Cat 1:
Elapidae: Dendroaspis angusticeps, Dendroaspis polylepis; Naja annulifera1, Naja
mossambica, Naja nigricollis; Viperidae: Bitis arietans
Cat 2:
Atractaspididae: Atractaspis bibronii; Colubridae: Dispholidus typus; Thelotornis
capensis, Thelotornis mossambicanus1; Elapidae: Naja melanoleuca; Viperidae:
Proatheris superciliaris
Mozambique:
Cat 1:
Elapidae: Dendroaspis angusticeps, Dendroaspis polylepis; Naja annulifera1, Naja
mossambica; Viperidae: Bitis arietans, Bitis gabonica1
Cat 2:
Atractaspididae: Atractaspis bibronii; Colubridae: Dispholidus typus; Thelotornis
capensis, Thelotornis mossambicanus1; Elapidae: Hemachatus haemachatus, Naja
melanoleuca; Viperidae: Proatheris superciliaris
Somalia:
Cat 1:
Elapidae: Dendroaspis polylepis; Naja ashei1 (south); Naja haje; Viperidae: Bitis
arietans; Echis pyramidum
Cat 2:
Atractaspididae: Atractaspis fallax; Colubridae: Dispholidus typus; Thelotornis
mossambicanus1; Elapidae: Naja pallida, Naja melanoleuca; Viperidae: Echis
hughesi (north)
The Sudan:
Cat 1:
Elapidae: Naja haje, Naja nigricollis; Viperidae: Bitis arietans; Echis pyramidum
Cat 2:
Atractaspididae: Atractaspis fallax, Atractaspis irregularis (south); Colubridae:
Dispholidus typus; Elapidae: Dendroaspis jamesoni (south), Dendroaspis polylepis
(south); Naja melanoleuca (south), Naja nubiae1, Naja pallida (south-east);
Viperidae: Bitis gabonica1 (south), Bitis nasicornis (south); Cerastes cerastes
Uganda:
Cat 1:
Elapidae: Naja ashei1 (north-east), Naja haje (north), Naja nigricollis; Dendroaspis
jamesoni, Dendroaspis polylepis; Viperidae: Bitis arietans, Bitis gabonica1
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Thelotornis
kirtlandii; Elapidae: Naja melanoleuca; Pseudohaje goldii; Viperidae: Atheris
squamigera; Bitis nasicornis
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
107
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
The United Republic of Tanzania:
Cat 1:
Elapidae: Dendroaspis angusticeps, Dendroaspis polylepis; Naja mossambica
(including Pemba Island), Naja nigricollis; Viperidae: Bitis arietans
Cat 2:
Atractaspididae: Atractaspis bibronii, Atractaspis fallax (north), Atractaspis
irregularis (north-east); Colubridae: Dispholidus typus; Thelotornis capensis,
Thelotornis kirtlandii (Mahali and Udzungwa Mountains), Thelotornis
mossambicanus1, Thelotornis usambaricus1 (East Usambara Mountains); Elapidae:
Naja ashei1 (reported in north-east), Naja annulata1, Naja haje (north), Naja
melanoleuca (west and coast, including Mafia Island), Naja pallida; Viperidae:
Atheris squamigera; Bitis gabonica1 (west and south-east), Bitis nasicornis (north);
Proatheris superciliaris
Zambia:
Elapidae: Dendroaspis polylepis; Naja anchietae1, Naja annulifera1, Naja
mossambica, Naja nigricollis; Viperidae: Bitis arietans, Bitis gabonica1
Atractaspididae: Atractaspis bibronii; Colubridae: Dispholidus typus; Thelotornis
capensis, Thelotornis kirtlandii, Thelotornis mossambicanus1; Elapidae: Naja
annulata1, Naja melanoleuca
Cat 1:
Cat 2:
South sub Saharan Africa
Botswana:
Elapidae: Dendroaspis polylepis; Naja anchietae1 (west), Naja annulifera1 (east),
Naja mossambica, Naja nivea (south-west); Viperidae: Bitis arietans
Atractaspididae: Atractaspis bibronii; Colubridae: Dispholidus typus; Thelotornis
capensis
Cat 1:
Cat 2:
Lesotho:
Elapidae: Naja nivea; Viperidae: Bitis arietans
Cat 1:
Cat 2:
Elapidae: Hemachatus haemachatus
Namibia:
Elapidae: Dendroaspis polylepis; Naja anchietae1, Naja nivea (central & southern),
Naja mossambica (north-east), Naja nigricincta1; Viperidae: Bitis arietans
Atractaspididae: Atractaspis bibronii; Colubridae: Dispholidus typus; Thelotornis
capensis; Elapidae: Naja nigricollis (Caprivi)
Cat 1:
Cat 2:
1
108
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
South Africa:
Elapidae: Dendroaspis angusticeps (Natal), Dendroaspis polylepis; Naja
annulifera11 (north-east), Naja nivea, Naja mossambica (north-east); Viperidae:
Bitis arietans
Atractaspididae: Atractaspis bibronii; Colubridae: Dispholidus typus; Thelotornis
capensis; Elapidae: Hemachatus haemachatus; Naja melanoleuca (Natal), Naja
nigricincta1 (north-west); Viperidae: Bitis gabonica1 (Natal);
Cat 1:
Cat 2:
Swaziland:
Elapidae: Dendroaspis polylepis; Naja annulifera1 Naja mossambica; Viperidae:
Bitis arietans
Atractaspididae: Atractaspis bibronii; Colubridae: Dispholidus typus; Thelotornis
capensis; Elapidae: Hemachatus haemachatus
Cat 1:
Cat 2:
Zimbabwe:
Elapidae: Dendroaspis polylepis; Naja anchietae1 (west), Naja annulifera1, Naja
mossambica; Viperidae: Bitis arietans
Atractaspididae: Atractaspis bibronii; Colubridae: Dispholidus typus; Thelotornis
capensis, Thelotornis mossambicanus1; Elapidae: Dendroaspis angusticeps (east);
Hemachatus haemachatus (Nyanga Mts); Naja melanoleuca (east); Viperidae: Bitis
gabonica1 (east)
Cat 1:
Cat 2:
West sub Saharan Africa
Benin:
Elapidae: Naja nigricollis; Viperidae: Bitis arietans; Echis ocellatus
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Elapidae:
Dendroaspis jamesoni; Naja katiensis, Naja melanoleuca, Naja senegalensis1;
Pseudohaje nigra; Viperidae: Bitis rhinoceros, Echis leucogaster (far north)
Cat 1:
Cat 2:
Burkina Faso:
Elapidae: Naja nigricollis, Naja katiensis; Viperidae: Bitis arietans; Echis ocellatus
Colubridae: Dispholidus typus; Elapidae: Dendroaspis polylepis; Naja melanoleuca,
Naja senegalensis1; Viperidae: Echis leucogaster
Cat 1:
Cat 2:
Cameroon:
Elapidae: Dendroaspis jamesoni; Naja haje, Naja nigricollis, Naja melanoleuca2;
Viperidae: Bitis arietans, Bitis gabonica1, Bitis nasicornis; Echis ocellatus
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Thelotornis
kirtlandii; Elapidae: Dendroaspis polylepis; Naja annulata1, Naja katiensis;
Pseudohaje goldii; Viperidae: Atheris broadleyi (East Province), Atheris squamigera
Cat 1:
Cat 2:
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
This large, highly venomous snake is common in forested areas of south-west Cameroon and a high
burden of injury may be expected, although clinical data with direct attribution are not yet available.
2
109
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Côte d’Ivoire:
Cat 1:
Elapidae: Dendroaspis viridis; Naja nigricollis, Naja melanoleuca, Naja
senegalensis1; Viperidae: Bitis arietans, Bitis nasicornis, Bitis rhinoceros1; Echis
ocellatus
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Thelotornis
kirtlandii; Elapidae: Dendroaspis polylepis; Naja katiensis; Pseudohaje goldii,
Pseudohaje nigra; Viperidae: Atheris chlorechis
The Gambia:
Cat 1:
Elapidae: Dendroaspis viridis; Naja nigricollis; Viperidae: Bitis arietans; Echis
jogeri
Cat 2:
Colubridae: Dispholidus typus; Elapidae: Naja katiensis, Naja melanoleuca, Naja
senegalensis1
Ghana:
Cat 1:
Elapidae: Dendroaspis viridis, Naja nigricollis, Naja senegalensis1; Viperidae: Bitis
arietans; Echis ocellatus
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus, Thelotornis
kirtlandii; Elapidae: Naja katiensis, Naja melanoleuca2; Pseudohaje goldii,
Pseudohaje nigra; Viperidae: Atheris chlorechis; Bitis nasicornis, Bitis rhinoceros1
Guinea:
Cat 1:
Elapidae: Dendroaspis polylepis, Dendroaspis viridis; Naja katiensis, Naja
nigricollis, Naja melanoleuca, Naja senegalensis1; Viperidae: Bitis arietans; Echis
jogeri
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Thelotornis
kirtlandii; Elapidae: Pseudohaje nigra; Viperidae: Atheris chlorechis; Bitis
nasicornis, Bitis rhinoceros1
1
2
110
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
The medical importance of this species may be higher in the forested zone of southern Ghana.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Guinea-Bissau:
Cat 1:
Elapidae: Dendroaspis viridis; Naja nigricollis, Naja melanoleuca, Naja
senegalensis1; Viperidae: Bitis arietans; Echis jogeri
Cat 2:
Colubridae: Dispholidus typus; Thelotornis kirtlandii; Viperidae: Bitis rhinoceros1
Liberia:
Cat 1:
Elapidae: Dendroaspis viridis; Naja melanoleuca, Naja nigricollis
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Thelotornis kirtlandii;
Elapidae: Pseudohaje nigra; Viperidae: Atheris chlorechis; Bitis nasicornis, Bitis
rhinoceros1
Mali:
Cat 1:
Elapidae: Naja katiensis, Naja nigricollis, Naja senegalensis1; Viperidae: Bitis
arietans; Echis jogeri (west); Echis leucogaster; Echis ocellatus
Cat 2:
Colubridae: Dispholidus typus; Elapidae: Naja melanoleuca; Viperidae: Cerastes
cerastes
Mauritania:
Cat 1:
Elapidae: Naja senegalensis1 (south-east); Viperidae: Cerastes cerastes; Echis
leucogaster
Cat 2:
Viperidae: Bitis arietans
The Niger:
Cat 1:
Elapidae: Naja nigricollis; Viperidae: Bitis arietans; Echis leucogaster, Echis
ocellatus
Cat 2:
Colubridae: Dispholidus typus; Elapidae: Naja haje (south-central), Naja katiensis,
Naja nubiae1; Naja senegelensis1 (south-west); Viperidae: Cerastes cerastes
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
111
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Nigeria:
Cat 1:
Elapidae: Dendroaspis jamesoni; Naja haje (north-east), Naja nigricollis;
Viperidae: Bitis arietans, Bitis gabonica1; Echis ocellatus
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Thelotornis
kirtlandii; Elapidae: Naja katiensis, Naja melanoleuca2; Naja senegalensis (northwest); Pseudohaje goldii, Pseudohaje nigra; Viperidae: Atheris squamigera; Bitis
nasicornis; Echis leucogaster (north)
Sao Tome and Principe:
Cat 1:
Elapidae: Dendroaspis jamesoni; Naja melanoleuca
Cat 2:
None
Senegal:
Cat 1:
Elapidae: Naja katiensis; Naja nigricollis; Viperidae: Bitis arietans; Echis
leucogaster; Echis jogeri
Cat 2:
Colubridae: Dispholidus typus; Elapidae: Dendroaspis polylepis; Dendroaspis
viridis; Naja melanoleuca, Naja senegalensis
Sierra Leone:
Cat 1:
Elapidae: Dendroaspis viridis; Naja nigricollis; Viperidae: Bitis arietans
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Thelotornis
kirtlandii; Elapidae: Naja melanoleuca3; Pseudohaje nigra; Viperidae: Atheris
chlorechis; Bitis nasicornis, Bitis rhinoceros1
Togo:
Cat 1:
Elapidae: Naja nigricollis, Naja senegalensis1; Viperidae: Bitis arietans (south);
Echis ocellatus
Cat 2:
Atractaspididae: Atractaspis irregularis; Colubridae: Dispholidus typus; Thelotornis
kirtlandii; Elapidae: Dendroaspis jamesoni, Dendroaspis viridis; Naja katiensis,
Naja melanoleuca; Pseudohaje goldii, Pseudohaje nigra; Viperidae: Atheris
chlorechis; Bitis nasicornis, Bitis rhinoceros1
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
The medical importance of this species may be higher in the southern rainforest belt of Nigeria, from
Ibadan in the west to Oban and Eket in the east, and in the forested southern quarter of Sierra Leone.
3
The medical importance of this species may be higher in the forested southern quarter of Sierra Leone.
2
112
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Middle East
Cyprus:
Cat 1:
None
Cat 2:
Viperidae: Macrovipera lebetina
Iran (Islamic Republic of):
Cat 1:
Elapidae: Naja oxiana; Viperidae: Echis carinatus; Macrovipera lebetina;
Pseudocerastes persicus
Cat 2:
Elapidae: Walterinnesia morgani1 (west); Viperidae: Eristicophis macmahonii (east);
Gloydius halys caucasicus; Montivipera raddei; Vipera spp.
Iraq:
Cat 1:
Viperidae: Echis carinatus; Macrovipera lebetina
Cat 2:
Elapidae: Walterinnesia morgani1; Viperidae: Cerastes gasperettii; Pseudocerastes
fieldi, Pseudocerastes persicus
Israel
Cat 1:
Viperidae: Daboia palaestinae1; Echis coloratus
Cat 2:
Atractaspididae: Atractaspis engaddensis; Elapidae: Walterinnesia aegyptia;
Viperidae: Cerastes cerastes, Cerastes gasperettii; Pseudocerastes fieldi
Jordan:
Cat 1:
Viperidae: Daboia palaestinae1; Echis coloratus
Cat 2:
Atractaspididae: Atractaspis engaddensis; Elapidae: Walterinnesia aegyptia;
Viperidae: Cerastes gasperettii; Macrovipera lebetina; Pseudocerastes fieldi
Kuwait and Qatar:
Cat 1:
Viperidae: Cerastes gasperettii
Cat 2:
Elapidae: Walterinnesia morgani1 (Kuwait)
Lebanon:
Cat 1:
Viperidae: Daboia palaestinae1; Macrovipera lebetina
Cat 2:
None
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
113
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Oman:
Cat 1:
Atractaspididae: Atractaspis andersonii (south-west); Viperidae: Bitis arietans
(south-west); Echis coloratus (south-west), Echis carinatus, Echis omanensis1
(north)
Cat 2:
Elapidae: Naja arabica1 (south-west); Viperidae: Cerastes gasperettii; Echis
khosatzkii (south-west); Pseudocerastes persicus
Saudi Arabia:
Cat 1:
Atractaspididae: Atractaspis andersonii (south-west); Viperidae: Cerastes
gasperettii; Echis coloratus, Echis borkini1 (south-west)
Cat 2:
Atractaspididae: Atractaspis engaddensis (north-west); Elapidae: Naja arabica
(south-west); Walterinnesia aegyptia (west), Walterinnesia morgani1 (central &
south); Viperidae: Bitis arietans (south-west); Cerastes cerastes (south-west);
Pseudocerastes fieldi
The Syrian Arab Republic:
Cat 1:
Viperidae: Daboia palaestinae1; Macrovipera lebetina
Cat 2:
Viperidae: Pseudocerastes fieldi
Turkey:
Cat 1:
Viperidae: Macrovipera lebetina; Montivipera xanthina1
Cat 2:
Elapidae: Walterinnesia morgani1 (south); Viperidae: Montivipera raddei1; Vipera
ammodytes; Vipera eriwanensis; Vipera spp.
The United Arab Emirates:
Cat 1:
Viperidae: Echis carinatus (east); Echis omanensis1
Cat 2:
Viperidae: Cerastes gasperettii; Pseudocerastes persicus
West Bank and Gaza Strip:
Cat 1:
Viperidae: Daboia palaestinae1; Echis coloratus
Cat 2:
Atractaspididae: Atractaspis engaddensis; Elapidae: Walterinnesia aegyptia;
Viperidae: Cerastes cerastes, Pseudocerastes fieldi
1
114
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Yemen:
Cat 1:
Atractaspididae: Atractaspis andersonii; Elapidae: Naja arabica1; Viperidae: Bitis
arietans; Echis borkini1, Echis coloratus
Cat 2:
Viperidae: Cerastes cerastes, Cerastes gasperettii; Echis khosatzkii
ASIA AND AUSTRALASIA
Central Asia
Armenia:
Cat 1:
Viperidae: Macrovipera lebetina;
Cat 2:
Viperidae: Montivipera raddei1; Vipera eriwanensis, Vipera spp.
Azerbaijan:
Cat 1:
Viperidae: Macrovipera lebetina
Cat 2:
Viperidae: Gloydius halys; Vipera eriwanensis; Vipera spp.
Georgia:
Cat 1:
Viperidae: Macrovipera lebetina; Vipera ammodytes
Cat 2:
Viperidae: Vipera renardi, Vipera ursinii, Vipera spp.
Kazakhstan, Kyrgyzstan, Tajikistan, Uzbekistan and Turkmenistan:
Cat 1:
Elapidae: Naja oxiana (except Kazakhstan & Kyrgyzstan); Viperidae: Echis
carinatus (except Kyrgyzstan); Macrovipera lebetina (except Kazakhstan &
Kyrgyzstan); Gloydius halys (throughout)
Cat 2:
Viperidae: Vipera renardi (except Turkmenistan)
Mongolia:
Cat 1:
Viperidae: Gloydius halys
Cat 2:
Viperidae: Vipera berus, Vipera renardi
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
115
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
East Asia
China:
China Mainland
Cat 1:
Elapidae: Bungarus multicinctus; Naja atra; Viperidae: Cryptelytrops albolabris1;
Daboia siamensis1; Deinagkistrodon acutus; Gloydius brevicaudus; Protobothrops
mucrosquamatus
Cat 2:
Colubridae: Rhabdophis tigrinus; Elapidae: Bungarus bungaroides (south-east
Tibet), Bungarus fasciatus; Naja kaouthia; Ophiophagus hannah; Viperidae:
Cryptelytrops septentrionalis (south Tibet); Gloydius halys, Gloydius intermedius1,
Gloydius ussuriensis; Himalayophis tibetanus (south Tibet); Protobothrops jerdonii,
Protobothrops kaulbacki, Protobothrops mangshanensis1; Vipera berus (Jilin,
western Xinjiang); Vipera renardi (western Xinjiang); Viridovipera stejnegeri1
Hong Kong, Special Administrative Region
Cat 1:
Elapidae: Bungarus multicinctus; Naja atra; Viperidae: Cryptelytrops albolabris1
Cat 2:
None
Taiwan Province
Cat 1:
Elapidae: Bungarus multicinctus; Naja atra; Viperidae: Protobothrops
mucrosquamatus; Viridovipera stejnegeri1
Cat 2:
Viperidae: Deinagkistrodon acutus; Daboia siamensis1
The Democratic People’s Republic of Korea:
Cat 1:
Viperidae: Gloydius brevicaudus
Cat 2:
Viperidae: Gloydius intermedius1, Gloydius ussuriensis; Vipera berus
Japan (including Ryukyu Islands):
Cat 1:
Viperidae: Gloydius blomhoffii (main islands); Protobothrops flavoviridis (Ryukyu
Islands)
Cat 2:
Colubridae: Rhabdophis tigrinus; Viperidae: Gloydius tsushimaensis (Tsushima);
Protobothrops elegans
The Republic of Korea:
Cat 1:
Viperidae: Gloydius brevicaudus
Cat 2:
Colubridae: Rhabdophis tigrinus; Viperidae: Gloydius intermedius1, Gloydius
ussuriensis
1
116
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
South Asia
Afghanistan:
Cat 1:
Elapidae: Naja oxiana; Viperidae: Echis carinatus; Macrovipera lebetina
Cat 2:
Elapidae: Bungarus caeruleus (east), Bungarus sindanus (east), Naja naja (reported
in south-east); Viperidae: Eristicophis macmahonii (south-west); Gloydius halys
(north)
Bangladesh:
Cat 1:
Elapidae: Bungarus caeruleus, Bungarus niger, Bungarus walli; Naja kaouthia;
Viperidae: Cryptelytrops erythrurus1
Cat 2:
Elapidae: Bungarus bungaroides, Bungarus fasciatus, Bungarus lividus; Naja naja;
Ophiophagus hannah; Viperidae: Cryptelytrops albolabris1 (far north-west); Daboia
russelii1 (west)
Bhutan:
Cat 1:
Elapidae: Bungarus niger; Naja naja
Cat 2:
Elapidae: Bungarus caeruleus, Bungarus fasciatus; Bungarus lividus; Naja kaouthia;
Ophiophagus hannah; Viperidae: Cryptelytrops erythrurus1; Daboia russelii1;
Protobothrops jerdonii
India:
Cat 1:
Elapidae: Bungarus caeruleus; Naja kaouthia (east), Naja naja (throughout);
Viperidae: Daboia russelii1; Echis carinatus; Hypnale hypnale (south-west)
Cat 2:
Elapidae: Bungarus bungaroides, Bungarus fasciatus, Bungarus lividus, Bungarus
niger, Bungarus sindanus, Bungarus walli; Naja oxiana (west), Naja sagittifera
(Andaman Islands); Ophiophagus hannah (south, north-east, Andaman Islands);
Viperidae: Cryptelytrops albolabris1, Cryptelytrops erythrurus1, Cryptelytrops
septentrionalis1; Gloydius himalayanus; Protobothrops jerdonii, Protobothrops
kaulbacki, Protobothrops mucrosquamatus; Trimeresurus gramineus (south India),
Trimeresurus malabaricus (south-west),
Nepal:
Cat 1:
Elapidae: Bungarus caeruleus, Bungarus niger; Naja naja, Naja kaouthia;
Viperidae: Daboia russelii1
Cat 2:
Elapidae: Bungarus bungaroides, Bungarus fasciatus; Bungarus lividus, Bungarus
walli; Ophiophagus hannah; Viperidae: Cryptelytrops septentrionalis1; Gloydius
himalayanus; Himalayophis tibetanus1; Protobothrops jerdonii;
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
117
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Pakistan:
Cat 1:
Elapidae: Bungarus caeruleus, Bungarus sindanus; Naja naja, Naja oxiana;
Viperidae: Daboia russelii1; Echis carinatus
Cat 2:
Viperidae: Eristicophis macmahonii (west); Gloydius himalayanus (north);
Macrovipera lebetina (west)
Sri Lanka:
Cat 1:
Elapidae: Bungarus caeruleus; Naja naja; Viperidae: Daboia russelii1; Hypnale
hypnale
Cat 2:
Elapidae: Bungarus ceylonicus; Viperidae: Echis carinatus; Hypnale nepa,
Trimeresurus trigonocephalus
South-east Asia
Brunei Darussalam:
Cat 1:
Elapidae: Naja sumatrana
Cat 2:
Elapidae: Bungarus fasciatus, Bungarus flaviceps; Calliophis bivirgatus, Calliophis
intestinalis; Ophiophagus hannah; Viperidae: Parias sumatranus1; Tropidolaemus
subannulatus
Cambodia:
Cat 1:
Elapidae: Bungarus candidus; Naja kaouthia, Naja siamensis; Viperidae:
Calloselasma rhodostoma; Cryptelytrops albolabris1; Daboia siamensis1
Cat 2:
Elapidae: Bungarus fasciatus, Bungarus flaviceps; Ophiophagus hannah; Viperidae:
Cryptelytrops macrops1
Indonesia (Sumatra, Java, Borneo, Sulawesi & Lesser Sunda Islands):
Cat 1:
Elapidae: Bungarus candidus (Sumatra & Java); Naja sputatrix (Java & Lesser
Sunda Islands), Naja sumatrana (Sumatra & Borneo); Viperidae: Calloselasma
rhodostoma (Java); Cryptelytrops albolabris1; Daboia siamensis1
Cat 2:
Elapidae: Bungarus fasciatus, Bungarus flaviceps (Sumatra & Borneo); Calliophis
bivirgatus, Calliophis intestinalis; Ophiophagus hannah (Sumatra, Borneo & Java);
Viperidae: Cryptelytrops insularis1, Cryptelytrops purpureomaculatus1 (Sumatra);
Parias sumatranus; Tropidolaemus subannulatus
1
118
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
The Lao People’s Democratic Republic:
Cat 1:
Elapidae: Bungarus candidus, Bungarus multicinctus; Naja atra (north), Naja
siamensis1 (south & east); Viperidae: Calloselasma rhodostoma; Cryptelytrops
albolabris1
Cat 2:
Elapidae: Bungarus fasciatus; Naja kaouthia (south & east); Ophiophagus hannah
Viperidae: Cryptelytrops macrops; Protobothrops jerdonii; Protobothrops
mucrosquamatus
Malaysia:
Cat 1:
Elapidae: Bungarus candidus (Peninsular Malaysia); Naja kaouthia (northern
Peninsular Malaysia), Naja sumatrana (Peninsular Malaysia, Sabah & Sarawak);
Viperidae: Calloselasma rhodostoma
Cat 2:
Elapidae: Bungarus fasciatus, Bungarus flaviceps; Calliophis bivirgatus; Calliophis
intestinalis; Ophiophagus hannah; Viperidae: Cryptelytrops purpureomaculatus1;
Parias sumatranus1; Tropidolaemus subannulatus
Myanmar:
Cat 1:
Elapidae: Bungarus magnimaculatus, Bungarus multicinctus; Naja kaouthia,
Naja mandalayensis; Viperidae: Cryptelytrops albolabris1, Cryptelytrops
erythrurus1; Daboia siamensis1
Cat 2:
Elapidae: Bungarus bungaroides (Kachin State), Bungarus candidus (Thaninthayi
Div.); Bungarus flaviceps (east Shan State), Bungarus niger (Chin State and Rakhine
State); Naja siamensis (adjacent Thailand border) Ophiophagus hannah; Viperidae:
Calloselasma rhodostoma (Thaninthayi Div.); Cryptelytrops purpureomaculatus;
Protobothrops jerdonii, Protobothrops kaulbacki, Protobothrops mucrosquamatus
(Kachin)
The Philippines:
Cat 1:
Elapidae: Naja philippinensis (Luzon), Naja samarensis (Mindanao), Naja
sumatrana (Palawan)
Cat 2:
Elapidae: Calliophis intestinalis; Ophiophagus hannah; Viperidae: Parias
flavomaculatus1; Tropidolaemus philippensis1; Tropidolaemus subannulatus1
Singapore:
Cat 1:
Elapidae: Bungarus candidus; Naja sumatrana
Cat 2:
Elapidae: Bungarus fasciatus; Calliophis bivirgatus, Calliophis intestinalis;
Viperidae: Cryptelytrops purpureomaculatus1
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
119
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Thailand:
Cat 1:
Elapidae: Bungarus candidus; Naja kaouthia, Naja siamensis1; Viperidae:
Calloselasma rhodostoma; Cryptelytrops albolabris1, Daboia siamensis1
Cat 2:
Elapidae: Bungarus fasciatus, Bungarus flaviceps; Calliophis bivirgatus, Calliophis
intestinalis; Naja sumatrana; Ophiophagus hannah; Viperidae: Cryptelytrops
macrops1; Parias sumatranus
Timor-Leste:
Cat 1:
Viperidae: Cryptelytrops insularis1
Cat 2:
Elapidae: Naja sputatrix (reported)
Viet Nam:
Cat 1:
Elapidae: Bungarus candidus, Bungarus multicinctus, Bungarus slowinskii
(north); Naja atra (north), Naja kaouthia (south); Viperidae: Calloselasma
rhodostoma; Cryptelytrops albolabris1 (throughout); Deinagkistrodon acutus
Cat 2:
Elapidae: Bungarus fasciatus, Bungarus flaviceps (south); Naja siamensis (south);
Ophiophagus hannah; Viperidae: Cryptelytrops macrops1; Protobothrops jerdonii,
Protobothrops mucrosquamatus (north); Viridovipera stejnegeri1
Australo-Papua (including Pacific Islands):
There are no medically important land snakes in American Samoa; Cook Islands; Fiji; French
Polynesia; Guam; Kiribati; Marshall Islands; Nauru; New Caledonia; New Zealand; Northern
Mariana Islands; Pitcairn Island; Samoa; Tokelau; Tonga; Tuvalu; or Wallis and Futuna Islands.
Fiji possesses a single terrestrial venomous snake species (Ogmodon vitianus) while the
Solomon Islands possess three terrestrial venomous species (Salomonelaps par; Loveridgelaps
elapoides and Parapistocalamus hedigeri) with no and few snakebites, respectively.
Australia:
Cat 1:
Elapidae: Notechis scutatus; Pseudechis australis2; Pseudonaja affinis,
Pseudonaja mengdeni1, Pseudonaja nuchalis, Pseudonaja textilis
Cat 2:
Elapidae: Acanthophis antarcticus, Acanthophis spp.; Austrelaps spp.;
Hoplocephalus spp.; Oxyuranus microlepidotus, Oxyuranus scutellatus, Oxyuranus
temporalis; Pseudechis spp.; Pseudonaja aspidorhyncha1, Pseudonaja spp.;
Tropidechis carinatus
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
Pseudechis australis is common and widespread and causes numerous snakebites; bites may be severe,
although this species has not caused a fatality in Australia since 1968.
2
120
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Indonesia (West Papua and Maluku):
Cat 1:
Elapidae: Acanthophis laevis1
Cat 2:
Elapidae: Acanthophis rugosus1; Micropechis ikaheka; Oxyuranus scutellatus;
Pseudechis papuanus, Pseudechis rossignolii1; Pseudonaja textilis
Papua New Guinea:
Cat 1:
Elapidae: Acanthophis laevis1; Oxyuranus scutellatus
Cat 2:
Elapidae: Acanthophis rugosus1; Micropechis ikaheka; Pseudonaja textilis;
Pseudechis papuanus, Pseudechis rossignolii1
EUROPE
There are no venomous snakes in Iceland, Ireland, Isle of Man, Outer Hebrides, Orkney or
Shetland Islands. Crete and most of the islands of the western Mediterranean are also without
venomous snakes.
Central Europe
Albania; Bosnia and Herzegovina; Bulgaria; Croatia; Romania; Serbia; Montenegro;
Slovenia; The former Yugoslav Republic of Macedonia:
Cat 1:
Viperidae: Vipera ammodytes
Cat 2:
Viperidae: Vipera berus, Vipera ursinii
The Czech Republic; Poland; Slovakia:
Cat 1:
None
Cat 2:
Viperidae: Vipera berus
Greece:
Cat 1:
Viperidae: Vipera ammodytes (including Corfu)
Cat 2:
Viperidae: Macrovipera schweizeri; Montivipera xanthina1; Vipera berus, Vipera
ursinii
Hungary:
Cat 1:
None
Cat 2:
Viperidae: Vipera berus; Vipera ursinii
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
121
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Eastern Europe
Belarus; Estonia; Latvia; Lithuania; The Republic of Moldova:
Cat 1:
None
Cat 2:
Viperidae: Vipera berus, Vipera nikolskii (Moldova), Vipera ursinii (Moldova)
The Russian Federation:
Cat 1:
Viperidae: Vipera berus
Cat 2:
Viperidae: Gloydius halys, Gloydius intermedius 1 , Gloydius ussuriensis; (far-east
Russia); Macrovipera lebetina (Dagestan); Vipera nikolskii; Vipera renardi, Vipera
spp.
Ukraine:
Cat 1:
None
Cat 2:
Viperidae: Vipera berus, Vipera nikolskii, Vipera renardi, Vipera ursinii
Western Europe
Austria:
Cat 1:
None
Cat 2:
Viperidae: Vipera ammodytes, Vipera berus
Belgium; Denmark; Finland; Germany; The Netherlands; Norway:
Cat 1:
None
Cat 2:
Viperidae: Vipera berus
France:
Cat 1:
Viperidae: Vipera aspis
Cat 2:
Viperidae: Vipera berus, Vipera ursinii
Italy:
Cat 1:
Viperidae: Vipera aspis
Cat 2:
Viperidae: Vipera ammodytes, Vipera berus, Vipera ursinii
Portugal:
Cat 1:
None
Cat 2:
Viperidae: Vipera latastei, Vipera seoanei
1
122
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Spain:
Cat 1:
None
Cat 2:
Viperidae: Vipera aspis, Vipera latastei, Vipera seoanei
Sweden; The United Kingdom of Great Britain and Northern Ireland:
Cat 1:
Viperidae: Vipera berus (not Northern Ireland)
Cat 2:
None
Switzerland:
Cat 1:
None
Cat 2:
Viperidae: Vipera aspis, Vipera berus
THE AMERICAS
North America
Canada:
Cat 1:
None
Cat 2:
Viperidae: Crotalus oreganus1, Crotalus viridis, Sistrurus catenatus
Mexico:
Cat 1:
Viperidae: Agkistrodon bilineatus, Agkistrodon taylori1; Crotalus atrox, Crotalus
scutulatus, Crotalus simus1, Crotalus totonacus1; Bothrops asper
Cat 2:
Elapidae: Micruroides euryxanthus, Micrurus nigrocinctus, Micrurus tener,
Micrurus spp.; Viperidae: Agkistrodon contortrix; Atropoides mexicanus, Atropoides
occiduus, Atropoides spp.; Bothriechis schlegelii, Bothriechis spp.; Cerrophidion
godmani, Cerrophidion spp.; Crotalus basiliscus, Crotalus molossus, Crotalus
oreganus1, Crotalus ruber, Crotalus tzabcan1, Crotalus viridis, Crotalus spp.;
Ophryacus spp.; Porthidium nasutum, Porthidium spp.; Sistrurus catenatus
The United States of America:
Cat 1:
Viperidae: Agkistrodon contortrix, Agkistrodon piscivorus; Crotalus adamanteus,
Crotalus atrox, Crotalus horridus, Crotalus oreganus1, Crotalus scutulatus,
Crotalus viridis
Cat 2:
Elapidae: Micrurus fulvius, Micrurus tener; Viperidae: Crotalus molossus, Crotalus
ruber, Crotalus spp., Sistrurus catenatus, Sistrurus miliarius
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
123
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Central America
The medically most important species are Bothrops asper and Crotalus simus11.
Belize:
Cat 1:
Viperidae: Bothrops asper
Cat 2:
Elapidae: Micrurus spp.; Viperidae: Agkistrodon bilineatus; Atropoides mexicanus;
Bothriechis schlegelii; Crotalus tzabcan1; Porthidium nasutum
Costa Rica:
Cat 1:
Viperidae: Bothrops asper; Crotalus simus1
Cat 2:
Elapidae: Micrurus nigrocinctus, Micrurus spp.; Viperidae: Agkistrodon bilineatus;
Atropoides mexicanus, Atropoides spp.; Bothriechis schlegelii, Bothriechis lateralis,
Bothriechis spp.; Cerrophidion godmani; Lachesis melanocephala, Lachesis
stenophrys; Porthidium nasutum, Porthidium ophrymegas, Porthidium spp.
El Salvador:
Cat 1:
Viperidae: Crotalus simus1
Cat 2:
Elapidae: Micrurus nigrocinctus; Micrurus spp.; Viperidae: Agkistrodon bilineatus;
Atropoides occiduus; Bothriechis spp.; Cerrophidion godmani; Porthidium
ophryomegas
Guatemala:
Cat 1:
Viperidae: Bothrops asper; Crotalus simus
Cat 2:
Elapidae: Micrurus nigrocinctus, Micrurus spp.; Viperidae: Agkistrodon bilineatus;
Atropoides mexicanus, Atropoides occiduus, Atropoides spp.; Bothriechis schlegelii,
Bothriechis spp.; Cerrophidion godmani; Crotalus tzabcan1, Porthidium nasutum,
Porthidium ophryomegas
Honduras:
Cat 1:
Viperidae: Bothrops asper
Cat 2:
Elapidae: Micrurus nigrocinctus, Micrurus spp.; Viperidae: Agkistrodon bilineatus;
Atropoides mexicanus, Atropoides occiduus, Atropoides spp.; Bothriechis marchi,
Bothriechis schlegelii, Bothriechis spp.; Cerrophidion godmani; Crotalus simus1;
Porthidium nasutum, Porthidium ophryomegas
1
124
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Nicaragua:
Cat 1:
Viperidae: Bothrops asper; Crotalus simus1
Cat 2:
Elapidae: Micrurus nigrocinctus, Micrurus spp.; Viperidae: Agkistrodon bilineatus;
Atropoides mexicanus; Bothriechis schlegelii; Cerrophidion godmani; Lachesis
stenophrys; Porthidium nasutum, Porthidium ophryomegas
Panama:
Cat 1:
Viperidae: Bothrops asper
Cat 2:
Elapidae: Micrurus mipartitus, Micrurus nigrocinctus, Micrurus spp.; Viperidae:
Atropoides mexicanus, Atropoides spp.; Bothriechis lateralis, Bothriechis schlegelii,
Bothriechis spp.; Cerrophidion godmani; Lachesis acrochorda, Lachesis stenophrys;
Porthidium nasutum, Porthidium lansbergii, Porthidium spp.
Caribbean
No medically important snakes occur naturally in Anguilla; Antigua and Barbuda; the Bahamas;
Barbados; Bermuda; The British Virgin Islands; Cayman Islands; Cuba; Dominica; the
Dominican Republic; Grenada; Guadeloupe; Haiti; Jamaica; Montserrat; the Netherlands
Antilles; Saint Kitts and Nevis; Saint Vincent and the Grenadines; and Turks and Caicos Islands.
Aruba; Martinique; Saint Lucia; Trinidad and Tobago, and offshore islands:
Cat 1:
Viperidae: Bothrops cf. atrox (Trinidad), Bothrops caribbaeus (St Lucia), Bothrops
lanceolatus (Martinique); Crotalus durissus (Aruba)
Cat 2:
Elapidae: Micrurus circinalis (Trinidad), Micrurus lemniscatus (Trinidad);
Viperidae: Lachesis muta (Trinidad)
South America
No venomous snakes are naturally occurring in the Falkland Islands; and no dangerously
venomous snakes are naturally occurring in Chile.
Argentina:
Cat 1:
Viperidae: Bothrops alternatus, Bothrops diporus1 Crotalus durissus
Cat 2:
Elapidae: Micrurus corallinus, Micrurus lemniscatus, Micrurus spp.; Viperidae:
Bothrops ammodytoides, Bothrops jararaca, Bothrops jararacussu, Bothrops
mattogrossensis, Bothrops neuwiedi, Bothrops pubescens
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
125
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Bolivia (Plurinational State of):
Cat 1:
Viperidae: Bothrops atrox, Bothrops mattogrossensis1 Crotalus durissus
Cat 2:
Elapidae: Micrurus lemniscatus, Micrurus spixii, Micrurus surinamensis, Micrurus
spp.; Viperidae: Bothrocophias hyoprora, Bothrocophias microphthalmus1; Bothrops
bilineatus, Bothrops brazili, Bothrops jararacussu, Bothrops jonathani, Bothrops
moojeni, Bothrops sanctaecrucis, Bothrops spp., Bothrops taeniatus; Lachesis muta
Brazil:
Cat 1:
Viperidae: Bothrops atrox, Bothrops jararaca, Bothrops jararacussu, Bothrops
leucurus, Bothrops moojeni; Crotalus durissus
Cat 2:
Elapidae: Micrurus corallinus, Micrurus lemniscatus, Micrurus spixii, Micrurus
surinamensis, Micrurus spp.; Viperidae: Bothrocophias hyoprora1, Bothrocophias
microphthalmus1, Bothrops alternatus, Bothrops bilineatus, Bothrops brazili,
Bothrops diporus, Bothrops mattogrossensis, Bothrops neuwiedi, Bothrops
pubescens, Bothrops taeniatus, Bothrops spp.; Lachesis muta
Colombia:
Cat 1:
Viperidae: Bothrops asper, Bothrops atrox, Bothrops bilineatus; Crotalus durissus
Cat 2:
Elapidae: Micrurus lemniscatus, Micrurus mipartitus, Micrurus nigrocinctus,
Micrurus spixii, Micrurus surinamensis, Micrurus spp.; Viperidae: Bothriechis
schlegelii; Bothrocophias hyoprora1, Bothrocophias microphthalmus1,
Bothrocophias spp.; Bothrops brazili, Bothrops taeniatus, Bothrops spp.; Lachesis
acrochorda1, Lachesis muta; Porthidium nasutum, Porthidium lansbergii
Ecuador:
Cat 1:
Viperidae: Bothrops asper, Bothrops atrox, Bothrops bilineatus; Lachesis muta
Cat 2:
Elapidae: Micrurus lemniscatus, Micrurus mipartitus, Micrurus spixii, Micrurus
surinamensis, Micrurus spp.; Viperidae: Bothriechis schlegelii; Bothrocophias
hyoprora1, Bothrocophias microphthalmus1, Bothrocophias spp.; Bothrops brazili,
Bothrops taeniatus, Bothrops spp.; Lachesis acrochorda1; Porthidium nasutum,
Porthidium spp.
French Guiana (France):
Cat 1:
Viperidae: Bothrops atrox, Bothrops brazili, Bothrops bilineatus; Crotalus durissus
Cat 2:
Elapidae: Micrurus lemniscatus, Micrurus surinamensis, Micrurus spp.; Viperidae:
Bothrops taeniatus; Lachesis muta
1
126
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Guyana:
Cat 1:
Viperidae: Bothrops atrox, Bothrops bilineatus, Bothrops brazili; Crotalus durissus
Cat 2:
Elapidae: Micrurus lemniscatus, Micrurus surinamensis, Micrurus spp.; Viperidae:
Bothrops taeniatus; Lachesis muta
Paraguay:
Cat 1:
Viperidae: Bothrops alternatus; Crotalus durissus
Cat 2:
Elapidae: Micrurus corallinus, Micrurus lemniscatus, Micrurus spixii, Micrurus
spp.; Viperidae: Bothrops diporus, Bothrops jararaca, Bothrops jararacussu,
Bothrops mattogrossensis, Bothrops moojeni, Bothrops neuwiedi, Bothrops spp.
Peru:
Cat 1:
Viperidae: Bothrops atrox, Bothrops bilineatus, Bothrops pictus; Crotalus
durissus; Lachesis muta
Cat 2:
Elapidae: Micrurus lemniscatus, Micrurus mipartitus, Micrurus spixii, Micrurus
surinamensis, Micrurus spp.; Viperidae: Bothriechis schlegelii; Bothrocophias
hyoprora, Bothrocophias microphthalmus; Bothrops asper; Bothrops brazili,
Bothrops mattogrossensis, Bothrops taeniatus, Bothrops spp.
Suriname:
Cat 1:
Viperidae: Bothrops atrox, Bothrops bilineatus, Bothrops brazili; Crotalus durissus
Cat 2:
Elapidae: Micrurus lemniscatus, Micrurus surinamensis, Micrurus spp.; Viperidae:
Bothrops taeniatus; Lachesis muta
Uruguay:
Cat 1:
Viperidae: Bothrops alternatus; Crotalus durissus
Cat 2:
Elapidae: Micrurus corallinus, Micrurus spp.; Viperidae: Bothrops pubescens1
Venezuela (Bolivarian Republic of):
Cat 1:
Viperidae: Bothrops atrox, Bothrops cf. atrox, Bothrops venezuelensis; Crotalus
durissus (including Isla de Margarita)
Cat 2:
Elapidae: Micrurus circinalis, Micrurus lemniscatus, Micrurus mipartitus, Micrurus
spixii, Micrurus surinamensis, Micrurus spp.; Viperidae: Bothriechis schlegelii;
Bothrops asper, Bothrops brazili, Bothrops bilineatus; Lachesis muta; Porthidium
lansbergii
1
Recent nomenclatural change. Refer to Tables 1 and 2 for details of previous names.
127
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
Herpetological references1
1. Ananjeva NB et al. The reptiles of Northern Eurasia. Sofia, Pensoft, 2006.
2. Ashton KG, de Querioz A. Molecular systematics of the western rattlesnake, Crotalus
viridis (Viperidae), with comments on the utility of the D-loop in phylogenetic studies of snakes.
Molecular Phylogenetics and Evolution, 2001, 21:176–189.
3. Babocsay G. A new species of saw-scaled viper of the Echis coloratus complex (Ophidia:
Viperidae) from Oman, Eastern Arabia. Systematics and Biodiversity, 2004, 1:503–514.
4. Basoglu M, Baran I. The reptiles of Turkey. Part II. The Snakes. Izmir, University of
Matbaasi, 1980.
5. Buys PJ, Buys PJC. Snakes of Namibia. Windhoek, Gamsberg Macmillan. 1983.
6. Bons J, Geniez P. Amphibiens et Reptiles du Maroc. Barcelona, Asociación Herpetológica
Española. 1996.
7. Boycott RC. A herpetological survey of Swaziland [MSc thesis]. Durban, University of
Natal, 1992.
8. Branch B. Field guide to the snakes and other reptiles of Southern Africa. Cape Town,
Sruik 1988.
9. Broadley DG et al. Snakes of Zambia: An atlas and field guide. Frankfurt am Main, Edition
Chimaira, 2003.
10. Broadley DG. FitzSimons snakes of Southern Africa. Cape Town, Delta Books, 1983.
11. Broadley DG. The herpetofaunas of the islands off the coast of south Moçambique.
Arnoldia Zimbabwe, 1990, 9:469–493.
12. Broadley DG. Reptiles and amphibians from the Bazaruto Archipelago, Mozambique.
Arnoldia Zimbabwe, 1992, 9:539–548.
13. Broadley DG. Review of the Dispholidini, with the description of a new genus and species
from Tanzania (Serpentes, Colubridae). Bulletin of the Natural History Museum London
(Zoology), 2002, 68:57–74.
14. Broadley DG. A review of the genus Thelotornis A. Smith in eastern Africa, with the
description of a new species from the Usambara Mountains (Serpentes: Colubridae:
Dispholidini). African Journal of Herpetology, 2001, 50:53–70.
15. Broadley DG. The herpetofaunas of the islands off the coast of south Moçambique.
Arnoldia Zimbabwe, 1990, 9:469–493.
16. Campbell JA, Lamar WW. The venomous snakes of the western hemisphere. Vols. I and II.
Ithaca, NY, Comstock-Cornell, 2004.
17. Creer S et al. Genetic and ecological correlates of intraspecific variation in pitviper venom
composition detected using matrix-assisted laser desorption time-of-flight mass spectrometry
(MALDI-TOF-MS) and isoelectric focusing. Journal of Molecular Evolution. 2003, 56:317–329.
1
128
Major regional guides have author names italicized.
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
18. da Silva VX. The Bothrops neuwiedi Complex. In: Campbell JA, Lamar WW (eds) The
venomous reptiles of the Western Hemisphere. Vol. I. Ithaca, NY, Comstock Publishing
2004:410–422.
19. David P, Ineich I. Les serpents venimeux du monde: systématique et répartition. Dumerilia,
1999, 3:3–499.
20. De Smedt J. The vipers of Europe. Hablech, JDS Verlag, 2001.
21. Dobiey M, Vogel G. Venomous snakes of Africa. Frankfurt am Main, Terralog Edition
Chimaira, 2006.
22. El Din SB. A guide to the reptiles and amphibians of Egypt. Cairo, American University of
Cairo Press, 2006.
23. Gasperetti J. Snakes of Arabia. Fauna of Saudi Arabia, 1988, 9:169–450.
24. Geniez P et al. The amphibians and reptiles of the Western Sahara. Frankfurt am Main,
Edition Chimaira, 2004.
25. Gloyd HK, Conant R. Snakes of the Agkistrodon complex. Oxford, OH, SSAR, 1990.
26. Guo P et al. New evidence on the phylogenetic position of the poorly known Asian pitviper
Protobothrops kaulbacki (Serpentes: Viperidae: Crotalinae) with a redescription of the species
and a revision of the genus Protobothrops. The Herpetological Journal 2007, 17:237–246.
27. Gumprecht A et al. Asian Pitvipers. Berlin, Geitje Books, 2004.
28. Gutberlet RL Jr, Campbell JA. Generic recognition of a neglected lineage of South
American pitvipers (Squamata: Viperidae: Crotalinae), with the description of a new species
from the Colombian Chocó. American Museum Novitates, 2001, 3316:1–15.
29. Khan MS. A guide to the snakes of Pakistan. Edition Chimaira, 2002.
30. Kreiner G. The snakes of Europe. Edition Chimaira, 2007.
31. Latifi M. The snakes of Iran. Oxford, OH, SSAR, 1985.
32. Le Berre M. Faune du Sahara 1: Poissons-amphibiens-reptiles. Editions Raymond
Chabaud, 1989.
33. Lenk P et al. Phylogeny and taxonomic subdivision of Bitis (Reptilia: Viperidae) based on
molecular evidence. In: Joger U (ed) Phylogeny and systematics of the Viperidae. Kaupia,
Darmstädter Beiträge zur Naturgeschichte, 1999, 8:31–38.
34. Lenk P et al. Evolutionary relationships among the true vipers (Reptilia: Viperidae) inferred
from mitochondrial DNA sequences. Molecular Phylogenetics and Evolution, 2001, 19:94–104.
35. Leviton AE et al. Handbook to Middle East amphibians and reptiles. Oxford, OH, SSAR,
1992.
36. Leviton AE et al. The dangerously venomous snakes of Myanmar. Illustrated checklist with
keys. Proceedings of the California Academy of Sciences, 2003, 54:407–462.
37. Malhotra A, Thorpe RS. A phylogeny of four mitochondrial gene regions suggests a
revised taxonomy for Asian pitvipers (Trimeresurus and Ovophis). Molecular Phylogenetics and
Evolution. 2004, 32:83–100.
129
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
38. McDiarmid RW et al. Snake species of the world: A taxonomic and geographical reference.
Vol.1. 1999.
39. Murphy JC. Amphibians and Reptiles of Trinidad and Tobago. Malabar, FL, Krieger, 1997.
40. Nilson G, Rastegar-Pouyani N. Walterinnesia aegyptia Lataste, 1887 (Ophidia: Elapidae)
and the status of Naja morgani Mocquard 1905. Russian Journal of Herpetology, 2007, 14:7–14.
41. Orlov NL, Barabanov AV. Analysis of nomenclature, classification, and distribution of the
Agkistrodon halys – intermedius complexes: a critical review. Russian Journal of Herpetology
1999, 6:167–192.
42. O’Shea M. A guide to the snakes of Papua New Guinea. Port Moresby, Independent
Publishing, 1996.
43. Parkinson CL et al. Phylogeography of the pitviper clade Agkistrodon: historical ecology,
species status, and conservation of cantils. Molecular Ecology, 2000, 9:411–420.
44. Pitman CRS. A guide to the snakes of Uganda, 2nd ed. Codicote, Wheldon & Wesley,
1974.
45. Quijada-Mascareñas JA et al. Phylogeographic patterns of trans-Amazonian vicariants and
Amazonian biogeography: the Neotropical rattlesnake (Crotalus durissus complex) as an
example. Journal of Biogeography, 2007, 34:1296–1312.
46. Roman B. Serpents de Haute-Volta. Ouagadougou, CNRST, 1980.
47. Schleich HH et al. Amphibians and reptiles of Nepal. Ruggall, Gantner Verlag, 2002.
48. Schleich HH et al. Amphibians and reptiles of North Africa. Koenigstein, Koeltz. 1996.
49. Spawls S, Branch B. The dangerous snakes of Africa. London, Blandford, 1995.
50. Spawls S, et al. A field guide to the reptiles of East Africa: Kenya, Tanzania, Uganda,
Rwanda and Burundi. London, Academic Press, Natural World, 2002.
51. Steward JW. The snakes of Europe. Newton Abbott David & Charles, 1971.
52. Street D. Reptiles of northern and central Europe. London, Batsford 1979.
53. Sweeney RCH. Snakes of Nyasaland. Amsterdam, Asher & Co, 1971.
54. Thorpe RS, Pook CE, Malhotra A. Phylogeography of the Russell's viper (Daboia russelii)
complex in relation to variation in the colour pattern and symptoms of envenoming. The
Herpetological Journal, 2007, 17:209–218.
55. Visser J, Chapman DS. Snakes and snakebite. Cape Town, Struik, 1978.
56. Vogel G. Venomous snakes of Asia. Frankfurt am Main, Terralog Edition Chimaira, 2006.
57. Vogel G et al. Revision of the Tropidolaemus wagleri-complex (Serpentes: Viperidae:
Crotalinae). I. Definition of included taxa and redescription of Tropidolaemus wagleri (Boie,
1827). Zootaxa, 2007, 1644:1–40.
58. Whitaker R, Captain A. Snakes of India: The field guide. Chennai, Draco Books, 2004.
130
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
59. Williams DJ et al. Origin of the eastern brownsnake, Pseudonaja textilis (Duméril, Bibron
and Duméril) (Serpentes: Elapidae: Hydrophiinae) in New Guinea: evidence of multiple
dispersals from Australia, and comments on the status of Pseudonaja textilis pughi Hoser 2003.
Zootaxa, 2008, 1703:47–61.
60. Williams DJ, Wüster W. Snakes of Papua New Guinea. In: Williams DJ et al (eds)
Venomous bites and stings in Papua New Guinea. Melbourne, AVRU University of Melbourne,
2005:33–64.
61. Wüster W et al. Snakes across the strait: trans-Torresian phylogenetic relationships in three
genera of Australo-Papuan snakes (Serpentes: Elapidae: Acanthophis, Oxyuranus and
Pseudechis). Molecular Phylogenetics and Evolution, 2005, 34:1–14.
62. Wuster W, Broadley DG. A new species of spitting cobra from north-eastern Africa
(Serpentes: Elapidae: Naja). Journal of Zoology, London, 2003, 259:345–359.
63. Wüster W, Broadley DG. Get an eyeful of this: a new species of giant spitting cobra from
eastern and north-eastern Africa (Squamata: Serpentes: Elapidae: Naja). Zootaxa, 2007,
1532:51–68.
64. Wüster W et al. Origin and evolution of the South American pitviper fauna: evidence from
mitochondrial DNA sequence analysis. In: Schuett GW, Höggren M, Douglas ME, Greene HW
(eds) Biology of the vipers. Eagle Mountain, Utah, Eagle Mountain Publishing, 2002:111–128.
65. Wüster W et al. Origin and phylogenetic position of the Lesser Antillean species of
Bothrops (Serpentes: Viperidae): biogeographical and medical implications. Bulletin of the
Natural History Museum London (Zoology), 2002, 68:101–106.
66. Wüster W et al. Redescription of Naja siamensis Laurenti, 1768 (Serpentes: Elapidae), a
widely overlooked spitting cobra from Southeast Asia: geographic variation, medical importance
and designation of a neotype. Journal of Zoology, 1997, 243:771–788.
67. Wüster W et al. Systematics of the Bothrops atrox species complex: insights from
multivariate analysis and mitochondrial DNA sequence information. In: Thorpe RS, Wüster W,
Malhotra A. (eds) Venomous snakes: ecology, evolution and snakebite. Oxford, Clarendon Press,
1997:99–113 (Symposia of the Zoological Society of London, No. 70).
68. Wüster W et al. The phylogeny of cobras inferred from mitochondrial DNA sequences:
evolution of venom spitting and the phylogeography of the African spitting cobras (Serpentes:
Elapidae: Naja nigricollis complex). Molecular Phylogenetics and Evolution, 2007, 45:437–453.
69. Wüster W et al. Synopsis of recent developments in venomous snake systematics. Toxicon,
1997, 35: 319–340.
70. Wüster W et al. Synopsis of recent developments in venomous snake systematics, No. 2.
Toxicon, 1998, 36:299–307.
71. Wüster W et al. Synopsis of recent developments in venomous snake systematics, No. 3.
Toxicon, 1999, 37:1123–1129.
72. Wüster W, McCarthy CJ. Venomous snake systematics: Implications for snake bite
treatment and toxinology. In: Bon C, Goyffon M (eds) Envenomings and their treatments. Lyon,
Fondation Mérieux, 1996:13–23.
73. Zhao E, Adler K. Herpetology of China. Oxford, OH, SSAR 1993.
131
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
© World Health Organization
APPENDIX 2
Summary protocol for manufacturing and control of snake antivenom
immunoglobulins
1. Antivenom batch information
a.
b.
c.
d.
e.
f.
g.
Name and address of manufacturer.............................................................
Batch number .............................................................................................
Date of filling ..............................................................................................
Liquid or freeze-dried .................................................................................
Expiry date ..................................................................................................
Number of vials or ampoules ......................................................................
Temperature of storage ...............................................................................
2. Control of the venom batch(es) used for animal immunization
a. Producer of venom and location .................................................................
b. Information on the snake contributing to the venom batch:
i. Scientific names of the snake species .............................................
ii. Number of snakes............................................................................
iii. Geographical origins of the snakes .................................................
c. Dates of collection of the venoms...............................................................
d. Expiry date of the venoms preparation .......................................................
e. Biochemical and biological characterization of the venoms ......................
Test performed ...................................................................................
Results................................................................................................
3. Control of plasma donor animals
a.
b.
c.
d.
e.
f.
Location of the animal herd ........................................................................
Animal species used for immunization.......................................................
Vaccinations performed ..............................................................................
Dates of animals immunization...................................................................
Control of antivenom antibody titre ...........................................................
Veterinary certificate of health of animal donor.........................................
4. Collection and storage of plasma
a.
b.
c.
d.
e.
f.
Method of collection ...................................................................................
Date of collection ........................................................................................
Date of storage ............................................................................................
Type of containers.......................................................................................
Temperature of storage ...............................................................................
Type and content of preservatives added (if any) .......................................
5. Transport of plasma to fractionation facility
a. Date of transport..........................................................................................
b. Temperature of transport.............................................................................
c. Date of arrival ............................................................................................
132
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
6. Plasma pooling and fractionation
a. Temperature of plasma storage at fractionation facility..............................
b. Volume of plasmas of different specificity pooled for the production of
polyspecific antivenoms (if applicable) ......................................................
c. Date of plasma pooling ...............................................................................
d. Volume of the manufacturing plasma pool .................................................
e. Number of animal donors contributing to the manufacturing plasma pool
f. Quality control of the manufacturing plasma pool
Test performed ...................................................................................
Results ................................................................................................
g. Type of active substance (intact IgG, fragments) .......................................
7. Preparation and control of final bulk
a. Volume of bulk antivenoms of different specificity pooled for the
production of polyspecific antivenoms (if applicable)................................
b. Concentration of preservatives (if used)
Type....................................................................................................
Method ...............................................................................................
Result..................................................................................................
c. Quality control of manufacturing plasma pool
Test performed ...................................................................................
Results ................................................................................................
8. Filling and containers
a.
b.
c.
d.
Date of filling ..............................................................................................
Quantity of containers .................................................................................
Volume of antivenoms per container ..........................................................
Date of freeze-drying (if any)......................................................................
9. Control tests on final product
a.
b.
c.
d.
Appearance..................................................................................................
Solubility (freeze-dried product).................................................................
Extractable volume......................................................................................
Venom-neutralizing potency test
Method ...............................................................................................
Venom used........................................................................................
Results ................................................................................................
e. Osmolality ...................................................................................................
f. Identity test
Method ...............................................................................................
Result..................................................................................................
g. Protein concentration
Method ...............................................................................................
Result..................................................................................................
133
WHO Guidelines for the Production, Control and Regulation of Snake Antivenom Immunoglobulins
h. Purity
Method ...............................................................................................
Result .................................................................................................
i. Molecular size distribution
Method ...............................................................................................
Result .................................................................................................
j. Test for pyrogens
Method ...............................................................................................
Result .................................................................................................
k. Sterility test
No. of containers examined ...............................................................
Method ...............................................................................................
Date at start of test .............................................................................
Date at end of test...............................................................................
l. Concentration of sodium chloride and other excipients
Method ...............................................................................................
Result .................................................................................................
m. Determination of pH
Result .................................................................................................
n. Concentration of preservatives (if used)
Type ...................................................................................................
Method ...............................................................................................
Result .................................................................................................
o. Chemical agents used in plasma fractionation
Type ...................................................................................................
Method ...............................................................................................
Result .................................................................................................
p. Inspection of final containers
Results................................................................................................
q. Residual moisture in freeze-dried antivenoms
Method ...............................................................................................
Result .................................................................................................
10. Internal certification
Certification by person taking overall responsibility for production of the antivenom
I certify that the batch No. ………………………. of snake antivenom immunoglobulin
satisfies the WHO Guidelines for the production, quality control and regulation of snake
antivenom immunoglobulins.
Signature…………………………………….......……..
Name (typed) …………………………………….........
Date…………………………………….......…………..
134
Cover by D.Meissner WHO/GRA
Download